Abstract
Background
Chronic lymphocytic leukaemia (CLL) is the most common cancer of the lymphatic system in Western countries. Several clinical and biological factors for CLL have been identified. However, it remains unclear which of the available prognostic models combining those factors can be used in clinical practice to predict long‐term outcome in people newly‐diagnosed with CLL.
Objectives
To identify, describe and appraise all prognostic models developed to predict overall survival (OS), progression‐free survival (PFS) or treatment‐free survival (TFS) in newly‐diagnosed (previously untreated) adults with CLL, and meta‐analyse their predictive performances.
Search methods
We searched MEDLINE (from January 1950 to June 2019 via Ovid), Embase (from 1974 to June 2019) and registries of ongoing trials (to 5 March 2020) for development and validation studies of prognostic models for untreated adults with CLL. In addition, we screened the reference lists and citation indices of included studies.
Selection criteria
We included all prognostic models developed for CLL which predict OS, PFS, or TFS, provided they combined prognostic factors known before treatment initiation, and any studies that tested the performance of these models in individuals other than the ones included in model development (i.e. 'external model validation studies'). We included studies of adults with confirmed B‐cell CLL who had not received treatment prior to the start of the study. We did not restrict the search based on study design.
Data collection and analysis
We developed a data extraction form to collect information based on the Checklist for Critical Appraisal and Data Extraction for Systematic Reviews of Prediction Modelling Studies (CHARMS). Independent pairs of review authors screened references, extracted data and assessed risk of bias according to the Prediction model Risk Of Bias ASsessment Tool (PROBAST). For models that were externally validated at least three times, we aimed to perform a quantitative meta‐analysis of their predictive performance, notably their calibration (proportion of people predicted to experience the outcome who do so) and discrimination (ability to differentiate between people with and without the event) using a random‐effects model. When a model categorised individuals into risk categories, we pooled outcome frequencies per risk group (low, intermediate, high and very high). We did not apply GRADE as guidance is not yet available for reviews of prognostic models.
Main results
From 52 eligible studies, we identified 12 externally validated models: six were developed for OS, one for PFS and five for TFS. In general, reporting of the studies was poor, especially predictive performance measures for calibration and discrimination; but also basic information, such as eligibility criteria and the recruitment period of participants was often missing. We rated almost all studies at high or unclear risk of bias according to PROBAST. Overall, the applicability of the models and their validation studies was low or unclear; the most common reasons were inappropriate handling of missing data and serious reporting deficiencies concerning eligibility criteria, recruitment period, observation time and prediction performance measures. We report the results for three models predicting OS, which had available data from more than three external validation studies:
CLL International Prognostic Index (CLL‐IPI)
This score includes five prognostic factors: age, clinical stage, IgHV mutational status, B2‐microglobulin and TP53 status. Calibration: for the low‐, intermediate‐ and high‐risk groups, the pooled five‐year survival per risk group from validation studies corresponded to the frequencies observed in the model development study. In the very high‐risk group, predicted survival from CLL‐IPI was lower than observed from external validation studies. Discrimination: the pooled c‐statistic of seven external validation studies (3307 participants, 917 events) was 0.72 (95% confidence interval (CI) 0.67 to 0.77). The 95% prediction interval (PI) of this model for the c‐statistic, which describes the expected interval for the model's discriminative ability in a new external validation study, ranged from 0.59 to 0.83.
Barcelona‐Brno score
Aimed at simplifying the CLL‐IPI, this score includes three prognostic factors: IgHV mutational status, del(17p) and del(11q). Calibration: for the low‐ and intermediate‐risk group, the pooled survival per risk group corresponded to the frequencies observed in the model development study, although the score seems to overestimate survival for the high‐risk group. Discrimination: the pooled c‐statistic of four external validation studies (1755 participants, 416 events) was 0.64 (95% CI 0.60 to 0.67); 95% PI 0.59 to 0.68.
MDACC 2007 index score
The authors presented two versions of this model including six prognostic factors to predict OS: age, B2‐microglobulin, absolute lymphocyte count, gender, clinical stage and number of nodal groups. Only one validation study was available for the more comprehensive version of the model, a formula with a nomogram, while seven studies (5127 participants, 994 events) validated the simplified version of the model, the index score. Calibration: for the low‐ and intermediate‐risk groups, the pooled survival per risk group corresponded to the frequencies observed in the model development study, although the score seems to overestimate survival for the high‐risk group. Discrimination: the pooled c‐statistic of the seven external validation studies for the index score was 0.65 (95% CI 0.60 to 0.70); 95% PI 0.51 to 0.77.
Authors' conclusions
Despite the large number of published studies of prognostic models for OS, PFS or TFS for newly‐diagnosed, untreated adults with CLL, only a minority of these (N = 12) have been externally validated for their respective primary outcome. Three models have undergone sufficient external validation to enable meta‐analysis of the model's ability to predict survival outcomes. Lack of reporting prevented us from summarising calibration as recommended. Of the three models, the CLL‐IPI shows the best discrimination, despite overestimation. However, performance of the models may change for individuals with CLL who receive improved treatment options, as the models included in this review were tested mostly on retrospective cohorts receiving a traditional treatment regimen. In conclusion, this review shows a clear need to improve the conducting and reporting of both prognostic model development and external validation studies. For prognostic models to be used as tools in clinical practice, the development of the models (and their subsequent validation studies) should adapt to include the latest therapy options to accurately predict performance. Adaptations should be timely.
Keywords: Adult; Female; Humans; Male; Age Factors; Bias; Biomarkers, Tumor; Calibration; Confidence Intervals; Discriminant Analysis; Disease-Free Survival; Genes, p53; Genes, p53/genetics; Immunoglobulin Heavy Chains; Immunoglobulin Heavy Chains/genetics; Immunoglobulin Variable Region; Immunoglobulin Variable Region/genetics; Leukemia, Lymphocytic, Chronic, B-Cell; Leukemia, Lymphocytic, Chronic, B-Cell/mortality; Leukemia, Lymphocytic, Chronic, B-Cell/pathology; Models, Theoretical; Neoplasm Staging; Prognosis; Progression-Free Survival; Receptors, Antigen, B-Cell; Receptors, Antigen, B-Cell/genetics; Reproducibility of Results; Tumor Suppressor Protein p53; Tumor Suppressor Protein p53/genetics
Plain language summary
How well do tools predict what happens with adults with newly‐diagnosed chronic lymphocytic leukaemia (CLL) over time?
What was the aim of this review?
There are many types of blood cancers called leukaemia. Chronic lymphocytic leukaemia (CLL) is the most common type. Twenty‐five per cent of people who have leukaemia have CLL. It is natural for people with newly‐diagnosed CLL and their families to want to know what will happen with their health in the future. They may be wondering if or when they will need treatment, if or when their disease will get worse or how long people live with CLL.
Researchers identified several characteristics that are associated with these outcomes. From these characteristics, they have tried to design tools that help predict what may happen to groups of people with newly‐diagnosed CLL.
The aim of this Cochrane Review is to evaluate and summarise those tools and studies that test the tools with other patient data.
What are the key messages from this review?
Reviewers found that there is no reliable way to predict what might happen over time to people who have (untreated) CLL. One reason is because the prediction tools have not been tested enough times with enough different people to know how well they really work.
Another reason is because researchers continue to develop more effective CLL treatment options that have better results, and the prediction tools have not kept up with advances in treatment.
What are the main results of the review?
We identified 52 tools that were designed to predict what may happen to people newly‐diagnosed with CLL. To find the best tools, we had to select the studies carefully. To apply these tools in clinical practice:
‐ a tool has to be tested by different researchers to predict what may happen with individuals with CLL in different geographic locations using different groups of people (i.e. age, gender, stage) with CLL. In other words, we would not include a tool if it was only tested on the people who provided their data to create it;
‐ the results of the tool should be consistent to prove that it works;
‐ the tests of the tool have to provide enough information to show how well the tool works. For example, the tests have to include large groups of people and enough information about the type of CLL they have.
We found three tools that met these requirements: the CLL International Prognostic Index (CLL‐IPI), the Barcelona‐Brno score, and the MDACC 2007 index score.
The CLL‐IPI did the best job at identifying people who would survive longer with CLL and people who would survive less long. However, we rated the quality of the CLL‐IPI studies as low because they did not provide all the information necessary to know how accurate the tool was. The Barcelona‐Brno score and the MDACC 2007 index score, tested on a smaller overall number of patients, showed lower discrimination between persons with a good as compared to a worse prognosis, and showed a similarly low quality of the studies.
Conclusion
More and better research is needed to develop and test the tools to help predict how CLL will behave for different groups of people over time. The tools must also adapt to accurately predict the performance of new treatments.
Summary of findings
Summary of findings 1. CLL International Prognostic Index.
| Prognostic models for chronic lymphocytic leukaemia in adult patients | ||||||||
|
Population: untreated individuals with CLL
Index model: CLL international prognostic index Timing: moment of prediction at diagnosis of CLL; moment of outcome occurrence not prespecified (any moment after diagnosis was included) Setting: inpatient and outpatient care | ||||||||
| Outcomes | Measure | № of participants (studies) | Summary measure | Pooled result (95% CI) | Comments | |||
| risk group | n (deaths) | pooled result (95% CI) | 95%‐PI | |||||
| Overall survival (OS)a | Discrimination | 7 studies 3307 patients 917 deaths |
c‐statistic of external validation studies | 0.72 (95% CI 0.67 to 0.77) | 0.59 to 0.83 | GRADEc | ||
| Calibrationb | 8 studies 4891 patients 875 deaths |
survival per risk group | low | 2497 (249) | 92.5% (89.2% to 94.8%) | 82.4% to 97.0% | Survival at 5 years GRADEc |
|
| intermediate | 1428 (233) | 85.0% (79.7% to 89.1%) | 68.8% to 93.6% | |||||
| high | 765 (280) | 64.9% (56.4% to 72.6%) | 41.9% to 82.6% | |||||
| very high | 201 (113) | 40.4% (29.3% to 52.6%) | 13.2% to 72.4% | |||||
| CI: confidence interval; PI: prediction interval | ||||||||
aThe CLL‐IPI was developed to predict overall survival. Although we identified external validation studies for other outcomes, we limited our analysis to the primary outcome of the development study. bNo calibration measures were reported, and so we used data on survival frequencies per group to compare expected versus observed survival – see Figure 1. cGRADE was not conducted, as currently, no GRADE guidance for prognostic models exists.
Summary of findings 2. Barcelona‐Brno model.
| Prognostic models for chronic lymphocytic leukaemia in adult patients | ||||||||
|
Population: untreated individuals with CLL
Index model: Barcelona‐Brno model Timing: moment of prediction at diagnosis of CLL; moment of outcome occurrence not prespecified (any moment after diagnosis was included) Setting: inpatient and outpatient care | ||||||||
| Outcomes | Measure | № of participants (studies) | Summary measure | Pooled result (95% CI) | Comments | |||
| risk group | n (deaths) | pooled result (95% CI) | 95% PI | |||||
| Overall survival (OS)a | Discrimination | 4 studies 1755 patients 416 deaths |
c‐statistic of external validation studies | 0.64 (95% CI 0.60 to 0.67) | 0.59 to 0.68 | GRADEc | ||
| Calibrationb | 3 studies 1974 patients 317 deaths |
survival per risk group | low | 1042 (88) | 90.5% (85.1% to 94.0%) | 80.4% to 95.7% | Survival at 5 years GRADEc |
|
| intermediate | 673 (131) | 79.7% (70.7% to 86.5%) | 63.1% to 90.0% | |||||
| high | 259 (98) | 62.5% (49.3% to 74.1%) | 41.3% to 79.7% | |||||
| CI: confidence interval; PI: prediction interval | ||||||||
aThe Barcelona‐Brno model was developed to predict overall survival. Although we identified external validation studies for other outcomes, we limited our analysis to the primary outcome of the development study. bNo calibration measures were reported, and so we used data on survival frequencies per group to compare expected versus observed survival – see Figure 2. cGRADE was not conducted, as currently, no GRADE guidance for prognostic models exists.
Summary of findings 3. MDACC 2007 index score.
| Prognostic models for chronic lymphocytic leukaemia in adult patients | ||||||||
|
Population: untreated individuals with CLL
Index model: MDACC 2007 index score Timing: moment of prediction at diagnosis of CLL; moment of outcome occurrence not prespecified (any moment after diagnosis was included) Setting: inpatient and outpatient care | ||||||||
| Outcomes | Measure | № of participants (studies) | Summary measure | Pooled result (95% CI) | Comments | |||
| Risk group | n (deaths) | pooled result (95% CI) | 95% PI | |||||
| Overall survival (OS)a | Discrimination | 7 studies 5127 patients 994 deaths |
c‐statistic of external validation studies | 0.65 (95% CI 0.60 to 0.70) | 0.51 to 0.77 | GRADEc | ||
| Calibrationb | 5 studies 3786 patients 511 deaths |
survival per risk group | low | 1202 (35) | 97.0% (94.3% to 98.4%) | 90.9% to 99.0% | Survival at 5 years GRADEc |
|
| intermediate | 2425 (393) | 82.3% (74.6% to 88.0%) | 61.5% to 93.1% | |||||
| high | 159 (83) | 45.6% (31.3% to 60.5%) | 21.2% to 72.3% | |||||
| CI: confidence interval; PI: prediction interval | ||||||||
aThe MDACC 2007 index score was developed to predict overall survival. Although we identified external validation studies for other outcomes, we limited our analysis to the primary outcome of the development study. bNo calibration measures were reported, and so we used data on survival frequencies per group to compare expected versus observed survival – see Figure 3. cGRADE was not conducted, as currently, no GRADE guidance for prognostic models exists.
Background
Description of the condition
Chronic lymphocytic leukaemia (CLL) is the most common form of malignant neoplasm (cancer) of the lymphatic system in Western countries. It is responsible for 25% of all leukaemias and occurs mainly in the elderly population (Chiorazzi 2005). The reported age‐adjusted incidence rate of CLL in the USA between 2012 and 2016 was 4.9 per 100,000 persons with an estimated 20,720 new cases and an age‐adjusted death rate of 1.2 per 100,000 persons per year (Howlader 2019). In the European Union, an estimated 46,000 individuals were living with CLL five years post‐diagnosis in 2006 (Watson 2008).
In CLL, mature B cells accumulate in the blood, bone marrow, lymph nodes and spleen (Herishanu 2013). The diagnosis of CLL is generally established based on blood counts, differential counts, blood smears and immunophenotyping (Hallek 2017). The requirement for a diagnosis of CLL has been modified from a chronic absolute lymphocytosis with more than 5.0 × 10⁹ cells per L to an absolute count of more than 5.0 × 10⁹ monoclonal B cells with CLL immunophenotype per L peripheral blood for the duration of at least three months (Hallek 2008; Hallek 2017). In the case of a monoclonal B‐cell count lower than 5.0 × 10⁹ per L and the absence of disease‐related symptoms, cytopenias or tissue involvement other than bone marrow, the condition is defined as monoclonal B‐lymphocytosis (Hallek 2008). A diagnosis of small lymphocytic lymphoma (SLL) is made when lymphadenopathy (enlarged lymph nodes) or splenomegaly (enlarged spleen) are caused by infiltrating CLL cells, and B lymphocytes in the peripheral blood do not exceed 5.0 × 10⁹ per L (Hallek 2008).
CLL is characterised by a highly variable clinical course and prognosis. Some individuals with CLL experience no or only few symptoms over many years, do not require treatment, and have a life expectancy comparable to that of a healthy individual. Other individuals already experience symptoms at diagnosis or shortly thereafter, and die within a few years despite treatment. The heterogeneity in clinical presentation makes it difficult for the physician to predict accurately whether a patient may benefit from an early, aggressive treatment strategy, and to provide the patient with relevant prognostic information.
The most commonly used early attempts to group individuals with CLL according to their risk are the two staging systems by Binet 1981 (Binet 1981) and Rai 1975 (Rai 1975), which distinguish between early (Rai 0; Binet A), intermediate (Rai I, II; Binet B) and advanced stages (Rai III, IV; Binet C). The disease stage is determined by the number of lymphocytes in the peripheral blood, presence of enlarged lymph nodes, presence of anaemia or thrombocytopenia (low platelet count), and presence of an enlarged liver or spleen. The prognostic value of the two staging systems is limited as survival times vary significantly within these stages. According to the guidelines by the European Society for Medical Oncology, B‐symptoms at diagnosis (fever, night sweats or weight loss) categorise people with early stage disease according to Binet or Rai, and are considered to have aggressive disease, requiring treatment (Eichhorst 2015).
Prognostic models
A prognostic model is a mathematical function that considers at least two prognostic factors, also called predictors, simultaneously with the aim to provide an estimate of an individual patient's probability to experience a certain health event within a defined time frame in the future (Alba 2017; Moons 2009; Riley 2019; Steyerberg 2013). Prognostic factors may be characteristics of the individual or the disease (e.g. age, gender, disease stage, biological or genetic information), which are likely to predict a patient‐relevant outcome, such as overall survival or disease progression (Riley 2013). By statistical modelling methods, these factors are often combined to form a weighted model able to accurately predict the likelihood of this outcome. Such a model aims at assisting the clinician to estimate the patient's prognosis and enhance shared decision‐making. Establishing a prognosis for the individual patient may also lead to risk‐stratified treatment recommendations (Alba 2017; Debray 2017; Moons 2009; Riley 2019; Steyerberg 2013).
To develop a prognostic model for a specific outcome, such as death or disease progression, various data sources such as cohort, nested case‐control or case‐cohort studies are considered appropriate, especially when data are prospectively collected. Data originating from randomised clinical trials, as a special form of prospectively collected data, can be used, but may limit generalisability due to more restrictive eligibility criteria, selective participation of specialised centres, trial effects and unrealistically precise predictor assessments (Collins 2015; Moons 2014; Moons 2019; Pajouheshnia 2019). Before a prognostic model is used in clinical practice, its predictive performance should be quantified. Apparent performance of the model is the model performance estimated from the same data as used for model development and usually provides an overly optimistic estimate due to overfitting of the model to this specific dataset. Internal validation tests the model performance in the development dataset by using techniques such as bootstrapping or cross‐validation. In order to test the generalisability of a prognostic model, predictive performance should be ideally assessed in several independent sets of data of individuals that were not used in the development and internal validation of the model, preferably by independent investigators to reduce bias. This process is called external validation (Moons 2015).
In the past few years, a number of clinical and biological CLL prognostic factors have been identified, including genomic aberrations, gene abnormalities (p53, ATM), mutation status of the variable segments of the immunoglobulin heavy chain genes (IgHV), or surrogate markers for these factors, such as CD38 and ZAP‐70 expression (Döhner 2000; El Rouby 1993; Kay 2007). Assessing recent molecular markers at time of diagnosis is expected to provide more reliable information regarding optimal time for treatment initiation, type of therapy and individual prognosis (Kay 2007; Shanafelt 2004; Wierda 2011; Zenz 2011). Thus, progressive and smouldering forms of the disease can now be separated more accurately than by using Rai or Binet staging systems alone. Moreover, the early recognition of aggressive stage A and indolent stage B and C disease would allow rational application of risk‐adapted treatment strategies. Factors influencing the choice of treatment include age, fitness to tolerate chemotherapy or immunotherapy or both, TP53 status, previous or current immune cytopenias, and evidence of lymphomatous transformation (Goede 2012).
The recent increase in availability of biological markers for CLL presents a challenge, as well as an opportunity to develop more precise prognostic models or algorithms that integrate a combination of markers and may guide counselling and treatment decisions for the individual patient. In order to identify the best‐performing tool to estimate prognosis for untreated individuals with CLL, a comprehensive evaluation of all currently available prognostic models and meta‐analysis of their predictive performance in external validation studies is urgently needed.
Health outcomes
The highly variable course of CLL and the possibility of having a normal life expectancy without progression or need for treatment entails that overall survival (OS) is one of the most important outcomes to be predicted by a prognostic model. In the USA, the median age at diagnosis is 70 years and the five‐year survival rate with CLL is 85.1% (Howlader 2019). Therefore, it is important to observe patients as long as possible to obtain a prognostic model that is meaningful not only for high‐risk individuals, but also for people with a less aggressive disease and longer survival.
As individuals with CLL are usually older, which implies an increased prevalence of comorbidities and decreased physical fitness, treatment may lead to serious adverse events and interactions with other medications. Hence, alternative meaningful outcomes to be predicted by a prognostic model include progression‐free survival (PFS) or treatment‐free survival (TFS, also sometimes referred to as time‐to‐first‐treatment). Treatment options for CLL have improved over time, thus, affecting survival rates but not the rates of treatment indication.
Why it is important to do this review
Although several prognostic factors have been identified during the last decade (Pflug 2014; Stilgenbauer 2014; Zaja 2013), they are controversial and there is no single prognostic factor available to determine treatment options in CLL patients. These factors have also been combined into numerous prognostic models. To date, no systematic review has been conducted to evaluate and assess the predictive performance of prognostic models in CLL, which would inform us which models have the greatest validity and therefore, would be preferred to guide clinical decision‐making. To shed light on this important research question, we conducted a systematic review and, where possible, a meta‐analysis of existing prognostic models for CLL and their corresponding validation studies.
Objectives
To identify, describe and appraise all prognostic models developed to predict overall survival (OS), progression‐free survival (PFS) or treatment‐free survival (TFS) in newly‐diagnosed (previously untreated) adults with CLL, and meta‐analyse their predictive performances.
Methods
Criteria for considering studies for this review
| Table 1. PICOTS system for prognostic models | ||
| P | Population | Untreated individuals with chronic lymphocytic leukaemia (CLL) at time of prediction |
| I | Index model(s) | All developed prognostic models for CLL and their corresponding external validation studies |
| C | Comparator | No predefined comparator |
| O | Outcome(s) | Overall survival (OS), progression‐free survival (PFS), treatment‐free survival (TFS) |
| T | Timing | Moment of prediction at diagnosis of CLL; moment of outcome occurrence not prespecified (any moment after diagnosis was included) |
| S | Setting | Not specified |
Types of studies
According to the checklist for critical appraisal and data extraction for systematic reviews of prediction modelling studies (CHARMS) (Moons 2014), we included:
prognostic model development studies without external validation in independent data;
prognostic model development studies with external validation in independent data reported in the same study; and
external model validation studies of previously reported models.
A prognostic model was defined as some form of mathematical function including at least two independent factors to predict OS, PFS or TFS. We included only models that combined prognostic factors known and assessed before treatment initiation. We excluded prognostic factor finding studies (i.e. studies aiming to establish one or several variables as independent prognostic factor(s) associated with an outcome, but not aiming to develop a model to be used for individualised predictions (i.e. to provide absolute prognostic outcome probabilities) in new patients, and model impact studies (i.e. studies aiming to investigate the impact of the use of a model in practice) (Bouwmeester 2012). We excluded studies that were published only as conference proceedings. We did not exclude studies with treatment during follow‐up time.
Participants
We included studies on individuals with previously untreated, confirmed B‐cell CLL. We included both male and female adults (age ≥ 18 years).
Types of outcomes to be predicted
Primary outcome
Models that predict OS as an outcome
We chose this outcome because it has the greatest clinical relevance and is most important for patients. Furthermore, death due to any cause is an objective endpoint not susceptible to bias of the outcome assessor. We did not require studies to have a minimum follow‐up time for inclusion in this review.
Secondary outcomes
Models that predict either PFS or TFS
We chose these outcomes as patients with similar survival may nevertheless have differing lengths of time without symptoms or need for treatment, depending both on initial treatment and disease characteristics. In case of immediate start of treatment, identification of patients with a lower probability to obtain a good response will help in making decisions regarding treatment, for example, deciding which patients might receive new or more aggressive therapy regimens. In case of a watch‐and‐wait strategy, differences in estimated prognosis can influence patient management regarding surveillance and treatment.
Search methods for identification of studies
Electronic searches
Reporting and therefore retrieval of prognostic model studies is very poor, as guidelines on reporting of prediction models have only recently been published (Collins 2015). For our first search, we did not use a specific search filter (Appendix 1). As this search strategy was not very specific, it yielded many results, which had to be screened in detail by two review authors. For the updated search, we integrated the search filter by Geersing 2012 to allow for a more specific strategy (Appendix 2).
We searched the following databases without applying any language restrictions in order to reduce language bias.
MEDLINE via Ovid (searched 24 June 2019; Appendix 1; Appendix 2).
Embase (searched 24 June 2019; Appendix 3).
We searched the following databases for ongoing trials.
ClinicalTrials.gov (clinicaltrials.gov; searched 5 March 2020; Appendix 4).
World Health Organization International Clinical Trials Registry Platform (WHO ICTRP) (searched 5 March 2020; Appendix 5).
Initially, we planned to include only models published after 1990. However, prognostic model studies are often conducted based on retrospective data, which in some cases included the analysis of blood samples frozen at diagnosis and analysed many years later. Thus, we decided not to limit our search strategy to date of publication. We did not screen conference proceedings, according to the section 'Types of prognostic models', where we stated that we would exclude conference abstracts based on limited information that would not allow us to assess risk of bias. In contrast to our planning (Skoetz 2016), we did not search the database of prognostic studies by the Prognostic Methods Group as it has not been developed. We did not search conference proceedings because of their limited information that would have complicated the inclusion and exclusion of studies as well as the risk of bias rating.
Searching other resources
As prespecified in the protocol (Skoetz 2016), we searched the following sources.
-
Handsearching of references
References of all identified trials, relevant review articles and current treatment guidelines for further literature.
-
Personal contacts
We contacted authors of relevant studies, study groups, experts and investigators from transplantation centres worldwide who are known to be active in the field for unpublished material or further information on ongoing studies.
In addition, and not reported in the protocol of this review, we tracked the citations of all included studies in October 2018 to identify possible additional validation studies of a developed model (Web of Science citation indexing).
Data collection and analysis
Selection of studies
During independent screening of titles and abstracts by two review authors (AA, LE, MT, NK, NS), we organised regular group discussions for congruency due to the novelty of the topic. The main discussion points can be found below. After title and abstract screening, full texts for all eligible studies were obtained and independently selected by two review authors (AA, LE, MT, NK, NS). Disagreements were solved by involving one or more additional review authors (Lefebvre 2019).
As recommended in the PRISMA statement (Moher 2009), we documented the total number of retrieved references and numbers of included and excluded studies in a flow chart.
Solving disagreements in study inclusion
The poor reporting and the fine line between the development of a prognostic model and prognostic factor (identification) studies made the screening of titles and abstracts and the final decision regarding the inclusion of studies challenging. During several group discussions, the following issues emerged repeatedly.
As we changed our pre‐planned limitation of the search to include studies from inception of the database, we obtained many studies labelled as "staging system." A staging system intends to describe the severity of a disease and, thus, indicate some prognostic and therapeutic information. Two staging systems for CLL, Rai and Binet, are commonly used in clinical practice. However, the predicted survival of individuals with the same disease stage can be highly variable. We decided to exclude staging systems, as they are not used by clinicians any longer (except for Rai and Binet). Instead, we focused on new algorithms with a high chance to include more recently identified prognostic factors.
We decided to exclude studies that build genetic signatures aiming to distinguish between good and poor prognosis of an individual, due to several reasons. First, the development of a genetic signature is often preceded by the identification of genetic markers on the same cohort from several hundred candidate markers, which introduces a high potential for overfitting of the algorithm to the development cohort (e.g. Houldsworth 2014). Second, genetic signatures can consist of a tremendous number of genes compared to the number of individuals included in the cohort (e.g. Ferreira 2014). Third, it is currently difficult to apply the algorithm of a genetic signature to an external cohort due to the complexity of the model.
The fine line between a multivariable model, which includes several prognostic factors to prove their independence from each other, and a prognostic model was not always evident from the abstract. When an abstract mentioned a multivariable model and the formation of risk groups, we considered the full text of the paper. We then excluded papers that did not proceed to build a prognostic model or score explicitly.
References were classified as an external validation study if the term "validation" was explicitly stated or the application of the prognostic model was clearly the main focus of the paper. We also included some publications where validation was not explicitly mentioned and performance measures were not reported, but where the authors put their focus on the application of one of the previously developed prognostic models (e.g. CLL‐IPI V ‐ Reda 2017 (Milano cohort); CLL‐IPI V ‐ Rigolin 2017 (Ferrera cohort)).
The decisions detailed above may seem subjective and were made in the case of this specific review. We would advise future authors who aim to evaluate prognostic models for a certain disease to clearly define the eligibility of different types of prognostic models at the protocol stage. This will help with objectivity throughout the screening process.
Data extraction and data management
Teams of two review authors (AA, LE, MT, NK, NS) independently extracted the data to enable assessment of applicability of the model for the review and the 'Risk of bias' assessment (see below). We contacted authors of individual studies for additional information, where required. We developed a standardised data extraction form containing the following items based on the CHARMS checklist (Moons 2014).
-
General information
Author, title, publication date, country, language, duplicate publications
-
Source of data
E.g. cohort, case‐control, randomised trial participants, or registry data
-
Participants
Participant eligibility and recruitment method (e.g. consecutive participants, location, number of centres, setting, inclusion and exclusion criteria)
Participant description
Details of treatments received
Study dates
-
Outcomes to be predicted
Definition and method for measurement of outcome
Was the same outcome definition (and method for measurement) used in all patients?
Was the outcome assessed without knowledge of the candidate predictors (i.e. blinded)?
Time of outcome occurrence or summary of duration of follow‐up
-
Candidate predictors
Number and type of predictors (e.g. demographics, patient history, physical examination, additional testing, disease characteristics, tumour markers)
Definition and method for measurement of candidate predictors
Timing of predictor measurement (e.g. at patient presentation, at diagnosis, at treatment initiation)
Were predictors assessed blinded for outcome, and for each other (if relevant)?
Handling of predictors in the modelling (e.g. continuous, linear, non‐linear transformations or categorised)
-
Sample size
Number of participants and number of outcomes/events
Number of outcomes/events in relation to the number of candidate predictors (events per variable)
-
Missing data
Number of participants with any missing value (include predictors and outcomes)
Number of participants with missing data for each predictor
Handling of missing data (e.g. complete‐case analysis, imputation, or other methods)
-
Model development
Modelling method (e.g. logistic, survival, neural networks, or machine learning techniques)
Modelling assumptions satisfied
Method for selection of predictors for inclusion in multivariable modelling (e.g. all candidate predictors, pre‐selection based on unadjusted association with the outcome)
Method for selection of predictors during multivariable modelling (e.g. full model approach, backward or forward selection) and criteria used (e.g. P value, Akaike Information Criterion)
Shrinkage of predictor weights or regression coefficients (e.g. no shrinkage, uniform shrinkage, penalised estimation)
-
Model performance
Calibration (calibration plot, calibration slope, Hosmer‐Lemeshow test) and discrimination (C‐statistic, D‐statistic, log‐rank) measures with confidence intervals
Classification measures (e.g. sensitivity, specificity, predictive values, net reclassification improvement) and whether a priori cut‐points were used
-
Model evaluation
Method used for testing model performance: development dataset only (random split of data, re‐sampling methods, e.g. bootstrap or cross‐validation, none) or separate external validation (e.g. temporal, geographical, different setting, different investigators)
In case of poor validation, whether model was adjusted or updated (e.g. intercept re‐calibrated, predictor effects adjusted, or new predictors added)
-
Results
Final and other multivariable models (e.g. basic, extended, simplified) presented, including predictor weights or regression coefficients, intercept, baseline survival, model performance measures (with standard errors or confidence intervals)
Any alternative presentation of the final prediction models, e.g. sum score, nomogram, score chart, predictions for specific risk subgroups with performance
Comparison of the distribution of predictors (including missing data) for development and validation datasets
-
Interpretation and discussion
Interpretation of presented models (confirmatory, i.e. model useful for practice versus exploratory, i.e. more research needed)
Comparison with other studies, discussion of generalisability, strengths and limitations
References of model development studies, for which no external validation studies have yet been published, are summarised in Studies awaiting classification. These studies fit the formal inclusion criteria of this review. However, they have not yet been tested in any additional cohort, which renders summary of performance in external cohorts impossible. They cannot yet be evaluated regarding their clinical usefulness, and are therefore not yet described in detail.
Risk of bias and applicability assessment
Since the publication of the review protocol, a 'Risk of bias' tool specifically designed for prognostic model studies (Prediction model Risk Of Bias ASsessment Tool, PROBAST) was published (Moons 2019; Wolff 2019). Instead of the CHARMS checklist for critical appraisal of prognostic modelling studies as reported in our protocol, we used this new tool recommended by the Cochrane Prognosis Methods Group to assess the risk of bias of the individual prediction models investigated in the included primary studies. Teams of two review authors (AA, LE, MT, NK, NS) independently assessed the risk of bias and applicability for each study. We assessed only validation studies with the outcome that a model was developed for, in line with meta‐analyses, hence PROBAST assessments reflect only the outcome of interest and not any additional outcomes reported in the same study.
After classifying each study into one of the three categories (model development with or without external validation in the same publication and external validation study of a previously developed model only), we assessed risk of bias according to the following four PROBAST domains (once per developed or validated model and per outcome).
Participants
Predictors
Outcome
Analysis
We answered signalling questions within each domain with one out of five options ('yes', 'probably yes', 'probably no', 'no', 'no information'); 'yes' always implied the absence of a potential bias‐generating aspect. We rated domain‐level 'Risk of bias' assessments using one of the following three options.
Low risk of bias: if the criterion is adequately fulfilled in the study, i.e. the study is at a low risk of bias for the given domain.
High risk of bias: if the criterion is not fulfilled in the study, i.e. the study is at high risk of bias for the given domain.
Unclear risk of bias: if the study report does not provide sufficient information to allow for a clear judgement or if the risk of bias is unknown for one of the domains listed above.
PROBAST additionally requires the judgement of the applicability of the model to the research question. The assessment occurs per domain (for the first three domains only) with the following response options: 'low concern regarding applicability', 'high concern regarding applicability' and 'unclear concern regarding applicability' (equivalent to the categories for risk of bias).
Based on these domain‐level judgements, we established an overall judgement per study based on PROBAST guidance for risk of bias:
Low risk of bias: if a prediction model evaluation is judged as low on all domains relating to bias or applicability.
High risk of bias: if an evaluation is judged as high for at least one domain.
Unclear risk of bias: if the prediction model evaluation is unclear in one or more domains and was rated as low in the remaining domains.
When information for a complete judgement was missing, we contacted the corresponding authors via email to request additional information to be able to make sound judgements on the 'Risk of bias' assessment. According to PROBAST, models that were developed, but not (yet) externally validated can be classified as 'high risk of bias', with the exception of extremely large samples (Wolff 2019). Due to the novelty of PROBAST and the subjectivity of ratings, we devoted a small section of the results to present our experience and agreements. Two authors of PROBAST (RW, KGMM) are also authors in our review and RW was involved in group discussions addressing disagreements between ratings.
Solving disagreements regarding PROBAST ratings
Due to the novelty of the 'Risk of bias' tool (PROBAST), we arranged frequent team meetings to discuss disagreements and discrepancies regarding ratings between the six authors who extracted data and assessed risk of bias and applicability. For reasons of transparency, we report the following challenges and our agreements.
The relevant observation time varies between diseases. For an indolent neoplasm like CLL, the clinicians in our team considered a median follow‐up of five years as appropriate for OS in a cohort with a normal case mix (i.e. no limitation to patients with late stage or high‐risk disease such as patients with del(17p) only).
The signalling question for the participant domain asks whether all inclusions and exclusions of participants were appropriate (item 1.2). Some publications listed missing values as part of the exclusion criteria. Missing values are also treated in the analysis domain (items 4.3 and 4.4). To avoid duplicate rating of the same issue, we decided to rate the risk of bias as high for the participant domain when complete data were required for eligibility, and rated the risk of bias as high for the analysis domain when the number and characteristics of the background sample were described but persons were excluded from analysis due to missing values.
Some publications did not report eligibility criteria and/or recruitment period. We rated these studies as unclear not only for risk of bias, but also for concerns of applicability, because we could not be sure if the included individuals matched our review question and a current application of the model.
When a publication lacked information necessary to answer a signalling question, but other available information could be used as an indication, we made assumptions regarding the answer (probably yes, probably no) to avoid the option 'no information'.
For the CLL‐IPI, we frequently encountered the problem that one predictor was replaced by a proxy predictor in the analysis of an external validation study. We assumed that this may happen in clinical practice, and included the external validation study to validate the CLL‐IPI, although strictly speaking, they did not apply the original model. We rated the concern for applicability as unclear in the domain predictor.
Some models developed to predict TFS include predictors also used for treatment indication, which means that the predictors are not excluded from the outcome definition (PROBAST item 3.3; e.g. O‐CLL1 model: Rai staging is included in the prognostic score, and is, in combination with other characteristics, a factor for treatment indication). This may lead to an overestimation of the association between predictor and outcome, which is called incorporation bias (Moons 2019). We answered the corresponding PROBAST item 3.3 with 'no' and considered the risk of bias to be high for the outcome domain. We decided that all corresponding external validations, which validate a model that include a predictor also used for treatment indication, are also considered to contain a high risk of bias.
The PROBAST guidance for item 4.1, which states that a study should include at least 20 events per candidate predictor for development studies and 100 events for validation studies is recommended for regression analysis with a binary outcome. The models included in our systematic review are mainly based on Cox proportional hazard models (Moons 2019). We therefore considered a cohort with more than 100 events to be an appropriate sample size for development and external validation samples (Collins 2016).
The testing of model assumptions (PROBAST item 4.6) was very rarely reported. We did not let this item influence our judgement regarding the analysis domain because, by default, all except one study would have been considered to be of unclear risk of bias. We assumed that authors tested the pre‐conditions before using their analysis type. However, we would advise authors of primary studies to improve reporting of the entire model development process.
Measures of prediction model performance
In contrast to 'classical' meta‐analysis focusing on treatment efficacy as the parameter of interest, there is no single recommended methodology to meta‐analyse the predictive performance of prognostic models (Debray 2012; Debray 2014). Hence, we extracted the reported predictive performance measures for each studied model from the development and validation studies, notably the model's discrimination and calibration in the development cohort (i.e. apparent performance, the performance measures after internal validation) and the performance found in a model's external validation.
Calibration refers to the accuracy of the predicted risk probabilities, which means the agreement between estimated and observed number of events in a cohort. A model might predict more events than the number of observed events, i.e. there is overestimation of the number of events. In contrast, a model can predict fewer events than observed meaning that the number of events is underestimated. In both situations, the model is miscalibrated. The use of a miscalibrated model in clinical practice can misguide clinical decisions based on such models (Van Calster 2019). Calibration can be presented as a calibration plot (expected probabilities plotted against observed outcome frequencies), as ratios between observed and expected number of events or outcome frequencies (O:E ratios) or calibration table (Debray 2017).
Discrimination refers to the ability of a prediction model to differentiate between those who do or do not experience the outcome event. A model has perfect discrimination if the predicted risks for all individuals who develop the outcome are higher than those for all individuals who do not experience the outcome. Discrimination is commonly estimated by the so‐called concordance index (c‐statistic, also sometimes called c‐index). The c‐statistic reflects the probability that for any randomly selected pair of individuals, one with and one without the outcome, the model assigns a higher probability to the individual with the outcome. The c‐index can range from 0 to 1, with 1 indicating perfect discriminative ability and 0.5 meaning the model's predictions equal chance. The c‐index is identical to the area under the receiver‐operating characteristic (ROC) curve for models with binary endpoints, and can be generalised for time‐to‐event (survival) models accounting for censoring (Debray 2017).
Both measures are needed to assess the performance of a prognostic model.
Unit of analysis issues
Ideally, a prognostic model is developed and tested on specially designed, prospectively followed cohort studies consisting of a representative case mix. However, more often we identified models or validations derived from retrospective data or data from randomised controlled trials (RCTs). Data from these populations are used for several purposes and models. Where a model was developed and externally validated in the same publication, we compared the institution and year of inclusion of individuals to assure independence of the cohorts. We also compared the institution and year of inclusion of all external validation cohorts belonging to one developed model to avoid overlaps in participants. When the same cohort was used to validate two or more different prognostic models, we considered them as separate studies. When RCT data were used, the randomisation of individuals was not considered by the authors of prognostic model studies.
Dealing with missing data
When data were missing, we requested additional information from the original investigators. When confidence intervals for measures of discrimination were still missing after we contacted the author, we calculated these according to a method proposed by Newcombe (Debray 2017; Debray 2018a; Newcombe 2006).
Investigation/description of heterogeneity
For the c‐statistic, we used the between‐study standard deviation (tau ‐ τ) to quantify possible heterogeneity. As we did not summarise O:E ratios to pool calibration, we merely visually inspected differences in survival per subgroup between cohorts. We planned to investigate and discuss clinical and statistical heterogeneity and design aspects of included studies mentioned in the section 'Data extraction and data management' based on subgroup analysis. We were unable to perform prespecified subgroup analysis based on diagnostic criteria due to the low number of studies with clear distinction between criteria.
Discussing reporting deficiencies
It is widely recommended that all prognostic models assess and report calibration and discrimination (Collins 2015; Moons 2015). However, it is known from numerous systematic reviews on methodological conduct and reporting of prognostic models in various disciplines that calibration is rarely reported and when it is reported (Heus 2018), it is done quite poorly. Hence, we also evaluated reporting deficiencies.
Methods and reporting in prognostic research often do not follow current methodological recommendations, limiting retrieval, reliability and applicability of these publications (Bouwmeester 2012; Peat 2014). There are some indications that prognosis research is cluttered with false‐positive studies which would not have been published if the results were negative. Moreover, studies evaluating development studies of prognostic models are not prospectively registered, and usually no protocol is published (Peat 2014). Therefore, it is difficult to assess publication bias. We used sensitive search strategies to increase retrieval (Geersing 2012).
Data synthesis
Data synthesis and meta‐analysis approaches
In the protocol, we explained that we would pool the performance measures for calibration and discrimination. During the review process however, new methodological developments became available and we chose to adopt them, as follows. Although we assessed both model development and external validations for risk of bias, we performed meta‐analysis only for validation studies, as there was only one development study per model.
When a particular model has been validated at least three times for the primary outcome, we applied a random‐effects model for pooling the logit transformation of the discrimination measure (the c‐statistic) using the meta‐analysis packages 'metamisc' and 'metafor' in the R statistical language (Debray 2014; Debray 2018b; Viechtbauer 2010).
We pooled only validation studies with the outcome that a model was developed for (e.g. we did not meta‐analyse validation studies that tested a model to predict TFS if the model was developed for OS).
We used random‐effects meta‐analysis, because validation studies typically differ regarding several parameters, including patient characteristics and design.
Where the c‐statistic was not reported, we did not estimate it. When the c‐statistic was provided without measures of uncertainties, we calculated the standard error and confidence intervals based on the P value or the combination of sample size and number of events, if available, according to Newcombe and colleagues (Debray 2017; Debray 2018a; Newcombe 2006).
We planned to summarise the measures of calibration. Unfortunately, calibration measures and the expected survival, both in development and external validation cohorts, were rarely reported. Instead, many studies reported the observed outcome frequency per subgroup at a specific time point. Though it is possible to estimate approximate O:E ratios from the reported observed survival per risk category and probabilities reported in the model development study (for scores), in collaboration with the Cochrane Prognosis Methods Group, we decided against this estimation because it is merely an approximate indication for the calibration of a model. For time‐to‐event outcomes, we would need to account for censoring (Debray 2018a), which was not possible based on the limited reporting of the studies. Instead, we illustrated the survival per risk category for each model with sufficient data graphically. We pooled survival per risk category for all external validations of one model and reported a prediction interval. The 95% prediction interval (95% PI) is an estimate of the range in which a future average survival frequency in a new validation study of the prognostic model will fall with a 95% probability.
Subgroup analysis
We planned to investigate whether the change of definition of CLL over time has affected the performance of a model (i.e. Cheson 1996; Hallek 2008). We did not perform this analysis because the number of studies that reported a clear distinction between the definitions was too low.
Sensitivity analysis
During the review process, and before data extraction and analysis, we decided to conduct sensitivity analysis based on the 'Risk of bias' rating, if a sufficient number of validation studies were available per model.
We decided post hoc to conduct the following model‐specific meta‐analyses.
Test the effect of studies that reported the area under the curve (AUC) instead of the c‐statistic (which we meta‐analysed together). These two measures for discrimination are the same for binary outcomes, but may differ for time‐to‐event data depending on the time point chosen to calculate the AUC.
Explore the effect of the estimation of the 95% CI for the c‐statistic. Estimating confidence intervals can introduce imprecision, therefore we aimed to explore the extend of the changes between reported and estimated 95% CIs.
For the CLL‐IPI, several external validation studies used a proxy prognostic factor for TP53 deletion or mutation, the cytogenetic aberration based on FISH, del(17p). Although we expected the effect to be small as the concordance between the two predictors is larger than 90% (Dicker 2009), using a proxy can reduce predictive performance.
Rating the certainty of evidence and summary of findings
Originally, we planned to use the most up‐to‐date GRADE guidance for this systematic review. However, no GRADE guidance for grading the certainty of results from meta‐analysis of prognostic models yet exists. Hence, for this review, we decided to refrain from applying GRADE.
Results
Description of studies
Results of the search
Our literature search (24 June 2019; MEDLINE and Embase) resulted in 22,857 potentially relevant references related to prognostic models for newly‐diagnosed untreated individuals with chronic lymphocytic leukaemia (CLL). Of these, we removed 4144 duplicates, leaving 18,713 references for title and abstract screening. We identified 284 full‐text references, which might fulfil our predefined inclusion criteria, including two references identified by reference and citation tracking. Of these, we excluded 214 references. We documented the overall number of screened, included and excluded references in a PRISMA flow diagram (Figure 4).
1.

Study flow diagram
This led to a total of 70 included references. Of these, 38 reported model developments only, six publications reported both model development and external validation(s), and 25 publications reported external validation only. In total, we identified 52 prognostic models (in 44 publications), and 35 external validation studies of included models that predicted the primary outcome of one of the included developed models (in 31 publications). Several of these publications offered secondary analyses with another outcome. These are only listed in this review if the primary outcome was not reported.
Out of the 52 developed models, 12 models were externally validated at least once either in the primary publication or as described in an additional publication (for an overview, see Appendix 6). Forty‐one models did not undergo any external validation.
The search of databases for ongoing trials resulted in 240 records (ClinicalTrials.gov: 209 and ICTRP: 31; 5 March 2020). Of these, 40 records were duplicates. We identified three records that referred to the potential use of developing a prognostic model. One cohort had already been included from the database search (O‐CLL‐GISL, development of a model and several validations, e.g. O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL)). One study is currently recruiting (NCT03436524), and one mentioned the aim to develop a staging system, however, it does not refer to any relevant publication yet (NCT00275054).
It is important to note the difference between 'reference' and 'study': one reference of a model development study may in addition to the model development also report one or several external validation studies of their model in independent cohorts. Furthermore, some references of external validations may report and compare the performances of several prognostic models in their cohort. Several publications used the same dataset to validate the same model(s).
References of model development studies without any external validation studies are summarised in Studies awaiting classification and Appendix 7.
Included studies
We described the characteristics of included studies per model and per predicted outcome. The numbers of external validations belonging to each developed model and outcome may not correspond to the number of included references. We defined a study as a validation of one model in one independent cohort, which implies that the validation of two different models in the same cohort are counted as two separate studies for the description in this review. In some publications, one model was externally validated in several cohorts, while other publications validated several models in one cohort. We presented the number of participants included in analysis instead of the number of participants in the study sample or complete registry as, often, missing values were reported to be an exclusion reason. We only included external validation studies that predicted the same outcome that the prognostic model was developed for.
Models with meta‐analysis (> 3 external validations)
CLL‐IPI (OS, CLL‐IPI D ‐ Bahlo 2016 (development cohort))
Barcelona‐Brno model (OS, Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort))
MDACC 2007 (OS, MDACC 2007 D ‐ Wierda 2007 (MDACC))
Models without meta‐analysis (1‐3 external validations)
GCLLSG model (OS, GCLLSG D ‐ Pflug 2014 (GCLLSG))
Rossi model (OS, Rossi D ‐ Rossi 2013 (Italian cohort))
Stephens (OS, Stephens OS D ‐ Stephens 2015 (Ohio cohort))
Baliakas model (TFS, Baliakas D ‐ Baliakas 2019 (multicentre))
GIMEMA model (PFS, GIMEMA D ‐ Molica 2005 (GIMEMA cohort))
MDACC 2011 (TFS, MDACC 2007 D ‐ Wierda 2007 (MDACC))
Morabito model (TFS, Morabito D ‐ Morabito 2009 (Italian cohort))
O‐CLL1 model (TFS, O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL))
Stephens model (TFS, Stephens TFS D ‐ Stephens 2015 (Ohio cohort))
Models with more than three external validation studies per outcome
Prognostic models for the prediction of OS (with meta‐analysis, > 3 external validations)
CLL‐IPI (CLL‐IPI D ‐ Bahlo 2016 (development cohort))
The CLL‐IPI model predicts OS and was derived from combined data from eight phase three RCTs conducted in multiple countries (France, Germany, Poland, UK, USA), with a total of 1799 individuals included for the model development. The recruitment period for the trials lasted from 1997 to 2009 (CLL‐IPI D ‐ Bahlo 2016 (development cohort)). The median follow‐up time was 79.9 months (interquartile range (IQR) 79.9 to 101.4 months). All disease stages were included; most individuals were classified as Rai I/II or Binet B stage (50% and 41% respectively). The final model includes five predictors: TP53 status, IgHV mutational status, serum B2‐microglobulin, clinical stage (Rai or Binet), and age. The original weighting of predictors was simplified to present as a risk score for easier use in clinical practice. We identified 11 external validation cohorts (CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014); CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort); CLL‐IPI V ‐ Da Cunha‐Bang 2016 (Danish cohort); CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort); CLL‐IPI V ‐ Gentile 2016 (Italian cohort); CLL‐IPI V ‐ Molica 2016 (O‐CLL1‐GISL); CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort); CLL‐IPI V ‐ Rani 2018 (Indian cohort); CLL‐IPI V ‐ Reda 2017 (Milano cohort); CLL‐IPI V ‐ Rigolin 2017 (Ferrera cohort); CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)), with 10 validations for OS and eight for TFS. The validation studies for OS included a total of 5485 individuals with available data presenting between 1983 and 2016. Two relevant validation studies lacked information regarding the recruitment period (CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort); CLL‐IPI V ‐ Rani 2018 (Indian cohort)). Due to lack of reporting, only 3307 individuals with 917 deaths from seven external validation studies were included in the meta‐analysis of the discrimination (CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014); CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort); CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort); CLL‐IPI V ‐ Gentile 2016 (Italian cohort); CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort); CLL‐IPI V ‐ Rani 2018 (Indian cohort); CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)). We provide a summary of the main characteristics of studies regarding the CLL‐IPI in Figure 5.
2.

CLL international prognostic index ‐ summary of characteristics of included studies
Barcelona‐Brno model (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort))
The Barcelona‐Brno model predicts OS and was derived from data from a retrospective, single‐centre cohort study conducted in Spain (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort)). TFS was reported as a secondary outcome. Model development included 524 individuals. The recruitment period was not reported. The median follow‐up time was 99.6 months, ranging from 1 to 456 months. All disease stages were included; most individuals were Rai 0 or Binet A stage (62% and 83% respectively). The model aimed to simplify the CLL‐IPI described below, therefore only combinations of the factors included in the CLL‐IPI were tested. The final model included three predictors: IgHV mutational status, and the genomic aberrations del(17p), and del(11q). We identified six external validation cohorts (Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort); Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo); Barcelona‐Brno V ‐ Molica 2017 (O‐CLL1‐GISL); Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.); Barcelona‐Brno V ‐ Rani 2018 (Indian cohort); Barcelona‐Brno V ‐ Reda 2017 (Milan cohort)), with five validations for OS and six for TFS. The validation studies for OS included a total of 2501 individuals presenting between 1983 and 2016. Two validation studies lacked information regarding the recruitment period (Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort); Barcelona‐Brno V ‐ Rani 2018 (Indian cohort)). Due to lack of reporting, only 1755 individuals with 416 deaths from three studies were included in meta‐analysis of discrimination (Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort); Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo); Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.); Barcelona‐Brno V ‐ Rani 2018 (Indian cohort)). We provide a summary of the main characteristics of studies regarding the Barcelona‐Brno model in Figure 6.
3.

Barcelona‐Brno score ‐ summary of characteristics of included studies
MDACC 2007 (Wierda 2007) (MDACC 2007 D ‐ Wierda 2007 (MDACC))
The MDACC 2007 model predicts OS and was derived from data from a retrospective, single‐centre cohort study conducted in the USA (MDACC 2007 D ‐ Wierda 2007 (MDACC)). A total of 1674 individuals were included for the model development. The recruitment period for the study lasted from 1981 to 2004. The median follow‐up time was 58.8 months (95% confidence interval (CI) 55.2 to 61.2 months). All disease stages were included; most individuals were classified as Rai I or Binet A stage (739/1674 and 1019/1674 respectively). The final model included six predictors: age, absolute lymphocyte count, serum B2‐microglobulin, nodal groups, Rai stage, and gender. The publication reports both the formula to be used in combination with a nomogram, and a simplified index score. We identified 10 external cohorts (MDACC 2007 V ‐ Trajkova 2013 (Macedonia); MDACC 2007 V ‐ Bulian 2011 (Italian‐Swiss); MDACC 2007 V ‐ Gentile 2014 (Italian cohort); MDACC 2007 V ‐ Gentile 2016 (Mayo cohort); MDACC 2007 V ‐ González Rodríguez (Cabueñes); MDACC 2007 V ‐ Molica 2010 (GIMEMA cohort); MDACC 2007 V ‐ Molica 2015 (O‐CLL1‐GISL); MDACC 2007 V ‐ Muñoz‐Novas 2018 (Spanish cohort); MDACC 2007 V ‐ Pflug 2014 (3 RCTs); MDACC 2007 V ‐ Rani 2018 (Indian cohort)), with eight validations for OS and six for TFS. Seven studies used the index score and three additionally calculated the formula to predict OS. For one study, it was unclear if the index score or the formula were used. The eight cohorts for which OS was predicted included 6928 individuals presenting between 1983 and 2013. One validation study lacked information regarding the recruitment period (MDACC 2007 V ‐ Rani 2018 (Indian cohort)). Only the simplified index score was sufficiently externally validated to be summarised in meta‐analysis. In total, 5127 individuals with 994 deaths from seven studies remained for inclusion in the meta‐analysis of discrimination (MDACC 2007 V ‐ Bulian 2011 (Italian‐Swiss); MDACC 2007 V ‐ Gentile 2014 (Italian cohort); MDACC 2007 V ‐ Gentile 2016 (Mayo cohort); MDACC 2007 V ‐ González Rodríguez (Cabueñes); MDACC 2007 V ‐ Muñoz‐Novas 2018 (Spanish cohort); MDACC 2007 V ‐ Pflug 2014 (3 RCTs); MDACC 2007 V ‐ Rani 2018 (Indian cohort)). We provide a summary of the main characteristics of studies regarding the MDACC 2007 model in Figure 7.
4.

MDACC 2007 index score ‐ summary of characteristics of included studies
Prognostic models for the prediction of PFS or TFS (with meta‐analysis, > 3 external validations)
We did not identify any prognostic model development studies that were externally validated more than three times for the outcomes PFS or TFS.
Models with one to three external validation studies per outcome
Prognostic models for the prediction of OS (without meta‐analysis, 1 to 3 external validations)
GCLLSG model (Pflug 2014) (GCLLSG D ‐ Pflug 2014 (GCLLSG))
The GCLLSG model predicts OS and was derived from combined data from three RCTs conducted in multiple countries (Australia, Austria, Belgium, Czech Republic, Denmark, France, Germany, Israel, Italy, New Zealand and Spain), with a total of 2007 individuals included for the model development (GCLLSG D ‐ Pflug 2014 (GCLLSG)). The recruitment period for the trials lasted from 1997 to 2006. The median follow‐up time was 63.4 months; ranges were not reported. All disease stages were included; most individuals were Rai II or Binet A stage (37.9% and 42.6%, respectively). The final model included eight predictors: gender, age, Eastern Co‐operative Oncology Group Performance Status (ECOG PS), del17p, del11q, IgHV mutational status, serum thymidine kinase, and serum B2‐microglobulin. We identified three external validation cohorts, with one validation for OS (676 individuals, GCLLSG V ‐ Pflug 2014 (Mayo cohort)), and two for TFS (GCLLSG V ‐ Molica 2015 (O‐CLL1‐GISL); GCLLSG V ‐ Rani 2018 (Indian cohort)). All validation studies lacked information regarding the recruitment period. We provide a summary of the main characteristics of studies regarding the GCLLSG model in Figure 8.
5.

GCLLSG model ‐ summary of characteristics of included studies
Rossi (Rossi 2013) (Rossi D ‐ Rossi 2013 (Italian cohort))
The Rossi model predicts OS and was derived from data from a retrospective, multicentre cohort study conducted in Italy (Rossi D ‐ Rossi 2013 (Italian cohort)). A total of 637 individuals were included for the model development. The recruitment period for the study lasted from 1996 to 2011. The median follow‐up time was 67.2 months; ranges were not reported. All disease stages were included; most individuals were classified as Rai 0 stage (74.9%). The final model included five predictors: TP53, BIRC3 DIS, SF3B1 M, NOTCH1 M, and del11q22‐q23. We identified two external validations (Rossi V ‐ Jeromin 2014 (Munich cohort); Rossi V ‐ Rossi 2013 (unclear)). One validation study included a total of 1160 individuals presenting between 2005 and 2010 (Rossi V ‐ Jeromin 2014 (Munich cohort)). The second validation study was conducted between 1996 and 2011; the authors failed to report the number of participants included in the validation cohort (Rossi V ‐ Rossi 2013 (unclear)). We provide a summary of the main characteristics of studies regarding the Rossi model in Figure 9.
6.

Rossi model ‐ summary of characteristics of included studies
Stephens model (Stephens 2015) (Stephens OS D ‐ Stephens 2015 (Ohio cohort))
Stephens and colleagues developed models for two outcomes, OS and TFS (Stephens OS D ‐ Stephens 2015 (Ohio cohort); Stephens TFS D ‐ Stephens 2015 (Ohio cohort)). They were derived from data from a retrospective, single‐centre cohort study conducted in the USA. A total of 114 individuals were included for the model development. The recruitment period for the study lasted from 2002 to 2012. The follow‐up time was not reported. All disease stages were included; most individuals were classified as Rai I/II stage (46%). The final models for both outcomes were simplified for use in clinical practice. The simplified risk score for OS includes three predictors: ECOG PS, age, and lactate dehydrogenase. The simplified risk score for TFS included five predictors: ECOG PS, Rai stage, age, white blood cell count, and del11q22.3. We identified one external validation study (Stephens OS V ‐ Stephens 2015 (MDACC)), which included 129 individuals. The recruitment period of the validation study was not reported. We provide a summary of the main characteristics of studies regarding the Stephens model in Figure 10.
7.

Stephens model for OS ‐ summary of characteristics of included studies
Prognostic models for the prediction of PFS or TFS (without meta‐analysis, 1 to 3 external validations)
Baliakas model (Baliakas 2019) (Baliakas D ‐ Baliakas 2019 (multicentre))
The Baliakas model predicts TFS and was derived from data from a retrospective, multicentre cohort study conducted in 10 European institutions (Baliakas D ‐ Baliakas 2019 (multicentre)). A total of 1900 individuals were included for the development of two separate models, dividing the study population into mutated (M‐CLL) and unmutated CLL (U‐CLL) patients. The recruitment period for the study was not reported. The median follow‐up time was 7.1 years, ranging from 0.1 to 33.1 years. Only individuals with disease stage Binet A were included. The final model for M‐CLL included three predictors: TP53 abnormality, trisomy 12, and stereotyped subset #2 defined as IGHV3‐21/IGLV3‐21 BcR IG. The final model for U‐CLL included three predictors: TP53 abnormality, del11q, and gender. We identified one external validation study (Baliakas V ‐ Baliakas 2019 (MLL + Scan.)), which included a total of 649 persons from two separate studies, one conducted at the Munich Leukemia Laboratory (508 persons) and one Scandinavian population‐based study (141 persons). We provide a summary of the main characteristics of studies regarding the Baliakas model in Figure 11.
8.

Baliakas model ‐ summary of characteristics of included studies
GIMEMA model (Molica 2005) (GIMEMA D ‐ Molica 2005 (GIMEMA cohort))
The GIMEMA model predicts PFS and was derived from data from a retrospective, multicentre cohort study conducted in Italy (GIMEMA D ‐ Molica 2005 (GIMEMA cohort)). A total of 1138 individuals were included for the model development. The recruitment period for the study lasted from 1991 to 2000. The median follow‐up time was 54 months, ranging from 4 to 309 months. Only individuals with disease stage Binet A were included. The final model included four predictors: lymphocyte doubling time, absolute peripheral blood lymphocytosis, Rai stage, and gender. We identified one external validation study (GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.)), which included 265 persons presenting between 1997 and 2007. We provide a summary of the main characteristics of studies regarding the GIMEMA model in Figure 12.
9.

GIMEMA model ‐ summary of characteristics of included studies
MDACC 2011 (Wierda 2011) (MDACC 2011 D ‐ Wierda 2011 (MDACC))
The MDACC 2011 model predicts TFS and was derived from data from a retrospective, single‐centre cohort study conducted in the USA (MDACC 2011 D ‐ Wierda 2011 (MDACC)). A total of 930 individuals were included for the model development. The recruitment period for the study lasted from 2004 to 2009. The median follow‐up time was 26 months, ranging from three to 73 months. All disease stages were included; most individuals were classified as Rai I stage (51%). The final model included six predictors: IgHV mutational status, diameter of largest palpated lymph node, FISH category (del11q or del17p versus none), number of involved lymph node sites, lactate dehydrogenase (LDH), and the IgHV mutational status and LDH interaction term. We identified one external validation study (MDACC 2011 V ‐ Molica 2016 (O‐CLL1‐GISL)), which included 328 persons presenting between 2006 and 2010. We provide a summary of the main characteristics of studies regarding the MDACC 2011 model in Figure 13.
10.

MDACC 2011 model ‐ summary of characteristics of included studies
Morabito model (Morabito 2009) (Morabito D ‐ Morabito 2009 (Italian cohort))
The Morabito model predicts TFS and was derived from data from a multicentre study conducted in Italy (Morabito D ‐ Morabito 2009 (Italian cohort)). The study design was not reported. A total of 262 individuals were included for the model development. The recruitment period was not reported. The median follow‐up time was 36 months, ranging from 12 to 180 months. Only individuals with disease stage Binet A were included. The final model included three predictors: IgHV mutational status, CD38, and ZAP‐70. We identified one external validation study (Morabito V ‐ Gentile 2014 (O‐CLL1‐GISL)), which included 480 persons presenting from 2007. There was no information regarding the end of the recruitment period. We provide a summary of the main characteristics of studies regarding the Morabito model in Figure 14.
11.

Morabito model ‐ summary of characteristics of included studies
O‐CLL1 model (Gentile 2016) (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL))
The O‐CLL1 model predicts TFS and was derived from data from a prospective, multicentre cohort study conducted in Italy (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL)). A total of 480 individuals were included for the model development. Recruitment for the study started in 2007; the end date was not reported. The median follow‐up time was 42 months, ranging from 6 to 82 months. Only individuals with disease stage Binet A were included. The final model included four predictors: Rai stage, absolute lymphocyte count (ALC), serum B2‐microglobulin, and IgHV mutational status. We identified two external validation cohorts for TFS (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort); O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)), which included a total of 626 persons. One validation study recruited participants between 2001 and 2008 (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort)); the other study lacked information regarding the recruitment period (O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)). We provide a summary of the main characteristics of studies regarding the O‐CLL1 model in Figure 15.
12.

O‐CLL‐1 model ‐ summary of characteristics of included studies
Stephens model (Stephens 2015) (Stephens TFS D ‐ Stephens 2015 (Ohio cohort))
For a description of the model for TFS in Stephens and colleagues (Stephens TFS D ‐ Stephens 2015 (Ohio cohort)), please see description in section OS (above), as the authors used the same development and validation cohorts for both outcomes. We provide a summary of the main characteristics of studies regarding the Stephens model in Figure 16.
13.

Stephens model for TFS ‐ summary of characteristics of included studies
Models without external validation studies
The 34 references with 41 model or score developments can be found in Studies awaiting classification, as it is widely recommended that developed prediction models should not be used in daily practice before they were validated at least once (Moons 2009; Moons 2015; Moons 2019; Steyerberg 2013). We have provided an overview of the main characteristics of these developed (but never validated) models in Appendix 7. In total, 19 models were developed to predict OS, 19 to predict TFS and three to predict PFS. These models were published between 1982 and 2019 and contained an average number of 3.8 predictors (range between 2 and 6). Nineteen models were derived from retrospective cohorts, three were on prospective cohorts and 19 did not report the study design clearly. The most commonly included predictors were IgHV status (mutated versus unmutated), B2‐microglobulin, age, clinical stage, genomic aberrations as defined by Döhner 2000, ZAP‐70 expression, CD38 expression and gender.
Excluded studies
During abstract screening, we excluded 18,713 references that clearly did not match our inclusion criteria. Of the remaining 283 full‐text references, we excluded 213 for the following reasons.
References of prognostic factor studies or prognostic factor identification studies (130 references) (Berke 2019; Bo 2014; Brejcha 2010; Brugiatelli 2007; Bulian 2014; Byrd 2006; Cailliod 2005; Callea 1999; Catovsky 1989; Cesano 2013; Chang 2003; Chauzeix 2018; Chen 1997; Chena 2008; Chevallier 2002; Chiaretti 2014; Christiansen 1994; Ciccone 2012; Claus 2012; Claus 2014; Cmunt 2002; Cocco 2005; Corcoran 2005; Cordone 1998; Cortese 2014; Coscia 2012; Crespo 2003; Cro 2009; D'Arena 2001; D'Arena 2007; Damle 1999; DeAndres‐Galiana 2016; Degan 2004; Delgado 2009; Delgado 2014; Del Guidice 2011; Del Poeta 2010; Del Principe 2004; Del Principe 2006; Di Raimondo 2001; Dong 2011; Dong 2014; Durak 2009; El‐Kinawy 2012; Gattei 2008; Gdynia 2018; Gentile 2016; Giudice 2018; Gogia 2014; Grabowski 2005; Han 1984; Hock 2010; Hus 2006; Jaksic 1981; Josefsson 2007; Juliusson 1986; Juliusson 1990; Kahraman 2014; Kardum‐Skelin 2008; Karmiris 1994; Khalifa 2002; Kim 2004; Kimby 1988; Knospe 1977; Koberda 1989; Korycka‐Wolowiec 2011; Krober 2002; Kryachok 2011; Kurec 1992; Lai 2002; Lech‐Maranda 2012; Lech‐Maranda 2013; Lecouvet 1997; Li 2008; Li 2017a; Lin 2002; Lin 2014; Lozano‐Santos 2014; Lucas 2015; Maffei 2007; Maffei 2010; Mansouri 2013; Marasca 2005; Marasca 2013; Martinelli 2008; Masic 1998; Mateva 2001; Matthews 2006; Matthews 2007; Matutes 2013; Miao 2018; Molica 1986; Molica 1988; Molica 1991; Molica 1994; Molica 1998; Molica 1999a; Molica 1999b; Molica 2008; Montserrat 1991; Morabito 2001; Morabito 2015a; Morabito 2018c; Morilla 2008; Nenova 2000; Nipp 2014; Nowakowski 2009; Nuckel 2006; Nuckel 2009; Ocana 2007; Oliveira 2011; Oscier 1990; Paolino 1984; Prokocimer 1985; Qin 2017; Resegotti 1989; Rissiek 2014; Ronchetti 2016; Sarmiento 2002; Shanafelt 2010; Spacek 2009; Stamatopoulos 2017; Strefford 2015; Szymczyk 2018; Vojdeman 2017; Vural 2014; Weiss 2011; Wierda 2003; Winkler 2010; Zenz 2009), and five references aimed at identifying prognostic factor thresholds (Dasgupta 2015; Davis 2016; Rossi 2010a; Tobin 2005a; Tobin 2005b).
The main focus was on the development of a diagnostic CLL staging system (21 references) (Apelgren 2006; Baccarani 1982; Binet 1981; Binet 1977; Chelazzi 1979; Ciocoiu 1988; De Faria 2000; De Rossi 1989; Ferrara 1981; Jaksic 1992; Molica 1984; Moreno 2019; Rai 1990; Rai 1975; Rossi 1986; Rozman 1979; Santoro 1979; Scolozzi 1981; Velardi 1980; Wu 2010; Zengin 1997).
Involved genetic analysis only (16 references), e.g. genetic subgrouping, genetic signature(s) or genetic clustering (Baliakas 2015; Bomben 2009; Bou Samra 2014; Chuang 2012; Ferreira 2014; Friedman 2009; Herold 2011; Houldsworth 2014; Morabito 2015b; Orgueira 2019; Queiros 2015; Raponi 2018; Rodriguez 2007; Vallat 2013; Van Damme 2012; Zucchetto 2006).
Included a population which did not match our PICOTS question (10 references), e.g. previously treated individuals with CLL (Giles 2003; Kardum‐Skelin 2009; Keating 2000; Krober 2006; Melo 1987; Nola 2004; O'Brien 1993; Rossi 2010b; Rossi 2011; Weinberg 2007).
Nine references focused on the characterisation of CLL and the prevalence of prognostic factors (Criel 1997; Cro 2010; Cuneo 2004; D'Arena 2012; Geisler 1997; Gonzalez 2013; Gonzalez‐Gascon 2015; Gonzalez‐Rodriguez 2010; Hallek 1999).
Nine references of other types of studies, e.g. risk factor study, diagnostic accuracy study (Cuneo 2018; Deslandes 2007; Dimier 2018; Fang 2019; Kay 2018; Kleinstern 2018; Plesingerova 2017; Salomon‐Nguyen 1995; Savvopoulos 2016).
Four references developed or used an outdated staging system, which is no longer used in today’s clinical practice (Bettini 1986; Chastang 1985; French Cooperative Group on CLL 1988; Mandelli 1987).
Five other types of references, e.g. review or comment to an excluded study (Bomben 2005; Grever 2006; Jaksic 2014; Matutes 2017; Nedeva 2018).
Three references evaluated outcomes not relevant to this review (Shanafelt 2017; Stamatopoulos 2009; Tallarico 2018).
Two references focused on scores or models which included predictors available at treatment initiation only (incorrect timing) (Gentile 2018; Nabhan 2017).
Reporting deficiencies
Appendix 8 describes the reporting deficiencies of the included studies per model for the primary outcome each model was developed for. The number of studies includes the development study. Data obtained by contacting authors are included as available.
For the Barcelona‐Brno model, the number of events for two studies and calibration for one study was obtained by contacting the corresponding authors of the primary studies. For the CLL‐IPI, information on total sample size and number of events was provided by one author. Information on calibration was not reported in publications at all, but was obtained by contacting the corresponding authors. For the GCLLSG model, the confidence intervals of the c‐statistic and calibration for the development study publication with one external validation was obtained from the authors. For the MDACC 2007 model, the c‐statistic and 95% CI for one external validation study was provided by the corresponding author. We did not obtain any additional information for the other models. We received primary data of one study, but were unable to reconstruct the analysis.
Of the 41 models without external validations, all studies reported the total number of individuals included for analysis. Eighteen studies lacked information on the recruitment period, 19 did not clearly report their study design, 24 did not report the calibration of their model and 31 did not report any measure of discrimination for their model (Appendix 7). Since we included studies with a wide range of publication years, the large amount of missing information may be a result of the methodological evolution of prognostic model research. Moreover, with our decision to also include scores where authors decided seemingly ad hoc to assign points and create a prognostic score, we have included studies that show more resemblance with prognostic factor studies (i.e. testing of the independence of factors from each other) than with a prognostic model development.
Summarised over all categories of included studies (development studies without external validations (N = 40), development studies with external validations (N = 12), external validations of primary outcome of an included model (N = 35)), information was especially lacking on calibration and discrimination. Calibration was reported in 17 and discrimination in 45 out of 87 studies.
Risk of bias and applicability assessment of included studies
Models with more than three external validation studies per outcome
Prognostic models for the prediction of OS (with meta‐analysis, > 3 external validations)
CLL‐IPI (CLL‐IPI D ‐ Bahlo 2016 (development cohort))
We rated the risk of bias of the development study as low for the domains: participants, predictors and outcome (CLL‐IPI D ‐ Bahlo 2016 (development cohort)), and high for the domain 'analysis' due to several reasons (univariable selection and dichotomisation of factors, missing data handling). We rated the risk of bias for the validation studies as low for the domain 'participants' for two studies (CLL‐IPI V ‐ Gentile 2016 (Italian cohort); CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort)), unclear in the case of six studies due to unclear eligibility criteria, and high for two studies due to inappropriate inclusion criteria (CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014); CLL‐IPI V ‐ Rani 2018 (Indian cohort)). We rated the risk of bias for the domains 'predictors' and 'outcome' as low for most studies, except one study with an unclear rating for the domain 'predictors' because predictor assessment has probably changed (CLL‐IPI V ‐ Gentile 2016 (Italian cohort)), and three studies with an unclear rating for the domain ‘'outcome' due to short observation time (CLL‐IPI V ‐ Da Cunha‐Bang 2016 (Danish cohort); CLL‐IPI V ‐ Rani 2018 (Indian cohort); CLL‐IPI V ‐ Rigolin 2017 (Ferrera cohort)). Concerning the domain 'analysis', we considered seven studies to have a high risk of bias due to inappropriate handling of missing data, low number of events and lack of reporting of performance measures. Three studies had a low rating (CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014); CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort); CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort)), and one had an unclear rating due to lack of performance measures (CLL‐IPI V ‐ Da Cunha‐Bang 2016 (Danish cohort)). One validation study did not examine the outcome of interest (CLL‐IPI V ‐ Molica 2016 (O‐CLL1‐GISL)) (Figure 17).
14.

Risk of bias (PROBAST) assessment of the CLL‐IPI model (Bahlo 2016)
We rated the concern for applicability of the development study as high for the domain 'participants' due to the high proportion of individuals with treatment indication (CLL‐IPI D ‐ Bahlo 2016 (development cohort)). We rated this low for the domains 'predictors' and 'outcome'. We rated concern for applicability as unclear for the domain 'participants' in all validation studies but two, which received a low rating (CLL‐IPI V ‐ Da Cunha‐Bang 2016 (Danish cohort); CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort)), due to unclear eligibility criteria. We rated concern for applicability as unclear for the domain 'predictors' in six validation studies because one predictor (TP53 mutation) was replaced by a proxy predictor (del(17p). The remaining four were rated as low (CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort); CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort); CLL‐IPI V ‐ Rigolin 2017 (Ferrera cohort); CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)). We considered the concern for applicability to be low for the domain 'outcome' in all validation studies. One validation study (CLL‐IPI V ‐ Molica 2016 (O‐CLL1‐GISL))) did not examine the outcome of interest (Figure 18).
15.

Applicability assessment for all developed models with external validations
Barcelona‐Brno model (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort))
We rated the risk of bias of the development study as unclear for the domain 'participants' due to missing information on eligibility criteria (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort)), low for the domains 'predictors' and 'outcome', and high for the domain 'analysis' due to the model‐building process (factor selection and weighting). We rated the risk of bias between the validation studies as unclear for the domain 'participants' in three studies (Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort); Barcelona‐Brno V ‐ Rani 2018 (Indian cohort); Barcelona‐Brno V ‐ Reda 2017 (Milan cohort)) due to missing eligibility criteria and recruitment period, and low in two studies (Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo); Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.)). We rated the risk of bias for the domains 'predictors' and 'outcome' as low for all validation studies except one (Barcelona‐Brno V ‐ Rani 2018 (Indian cohort)), which we rated as unclear for the domain 'outcome' due to the short observation time. Concerning the domain 'analysis', we considered all studies to be at high risk of bias (especially due to handling missing data, low number of events and reporting of performance measures) except one (Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort)), which we rated as unclear. One validation study did not examine the outcome of interest (Barcelona‐Brno V ‐ Molica 2017 (O‐CLL1‐GISL)) (Figure 19).
16.

Risk of bias (PROBAST) assessment of the Barcelona‐Brno model (Delgado 2017)
We rated the concern for applicability of the development study as unclear for the domain 'participants' and low for the domains 'predictors' and 'outcome' (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort)). Similarly, we rated concern for applicability as unclear for the domain 'participants', and low for the domains 'predictors' and 'outcome' in three validation studies (Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort); Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo); Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.)) due to undefined eligibility criteria. We assigned two validation studies (Barcelona‐Brno V ‐ Rani 2018 (Indian cohort); Barcelona‐Brno V ‐ Reda 2017 (Milan cohort)) the rating unclear for the domain 'predictors' due to lack of information on the timing of predictor assessment. One validation study did not examine the outcome of interest (Barcelona‐Brno V ‐ Molica 2017 (O‐CLL1‐GISL)) (Figure 18).
MDACC 2007 (Wierda 2007) (MDACC 2007 D ‐ Wierda 2007 (MDACC))
We rated the risk of bias for the development study as low across all domains (MDACC 2007 D ‐ Wierda 2007 (MDACC)). We rated the risk of bias across the validation studies as low for the domain 'participants' in six validation studies and high in three validation studies (MDACC 2007 V ‐ Gentile 2016 (Mayo cohort); MDACC 2007 V ‐ Rani 2018 (Indian cohort); MDACC 2007 V ‐ Trajkova 2013 (Macedonia)) due to unclear eligibility criteria. We considered all validation studies to have a low risk of bias for the domains 'predictors'. We rated three studies as unclear for the domain 'outcome' (MDACC 2007 V ‐ Molica 2010 (GIMEMA cohort); MDACC 2007 V ‐ Rani 2018 (Indian cohort); MDACC 2007 V ‐ Trajkova 2013 (Macedonia)) because of the short observation time. Concerning the domain 'analysis', we considered all validation studies to have a high or unclear risk of bias due to the low number of events and inappropriate handling of missing data. One validation study did not examine the outcome of interest (MDACC 2007 V ‐ Molica 2015 (O‐CLL1‐GISL)) (Figure 20).
17.

Risk of bias (PROBAST) assessment of the MDACC 2007 model (Wierda 2007)
We rated the concern for applicability of the development study as low across all domains (MDACC 2007 D ‐ Wierda 2007 (MDACC)). We rated concern for applicability as low for the domain 'participants' in five validation studies and unclear in four validation studies due to unclear eligibility criteria (MDACC 2007 V ‐ Gentile 2014 (Italian cohort); MDACC 2007 V ‐ Gentile 2016 (Mayo cohort); MDACC 2007 V ‐ Pflug 2014 (3 RCTs); MDACC 2007 V ‐ Rani 2018 (Indian cohort)). Concerning the domain 'predictors', we considered the concern for applicability to be low in seven validation studies, high in one study (MDACC 2007 V ‐ Molica 2010 (GIMEMA cohort)), and unclear in one study (MDACC 2007 V ‐ Rani 2018 (Indian cohort)). Concern for applicability was rated as low for the domain 'outcome' in all validation studies except one, for which we assigned a high rating (MDACC 2007 V ‐ Molica 2010 (GIMEMA cohort)). One validation study did not examine the outcome of interest (MDACC 2007 V ‐ Molica 2015 (O‐CLL1‐GISL)) (Figure 18).
Prognostic models for the prediction of PFS or TFS (with meta‐analysis, > 3 external validations)
We did not identify any prognostic model development studies that were externally validated more than three times for the outcomes PFS, TFS or TFS.
Models with one to three external validation studies per outcome
Prognostic models for the prediction of OS (without meta‐analysis, 1 to 3 external validations)
GCLLSG model (Pflug 2014) (GCLLSG D ‐ Pflug 2014 (GCLLSG))
We rated the risk of bias for the development study as low for all domains except 'analysis' (GCLLSG D ‐ Pflug 2014 (GCLLSG)). We rated 'analysis' at high risk of bias due to dichotomisation and univariable selection of factors, inappropriate handling of missing data and simplification of the model (loss of original weighting). We considered the only validation study with a matching outcome to have a high risk of bias for the domains 'participants' and 'analysis' due to inclusion of participants with available data and low number of events (GCLLSG V ‐ Pflug 2014 (Mayo cohort)). Risk of bias was low for the domains 'predictors' and 'outcome'. The other two validation studies did not examine the outcome of interest (GCLLSG V ‐ Molica 2015 (O‐CLL1‐GISL); GCLLSG V ‐ Rani 2018 (Indian cohort)) (Figure 21).
18.

Risk of bias (PROBAST) assessment of other models that were externally validated
We rated the concern for applicability of the development study as unclear for the domain 'participants' because the sample was based on RCT data that may not be representative for all individuals with CLL (GCLLSG D ‐ Pflug 2014 (GCLLSG)). Concern was low for the domains 'predictors' and 'outcome'. Similarly, we rated concern for applicability of the validation study as unclear for the domain 'participants' due to eligibility criteria and low for the domains 'predictors' and 'outcome' (GCLLSG V ‐ Pflug 2014 (Mayo cohort)). The other two validation studies did not examine the outcome of interest (GCLLSG V ‐ Molica 2015 (O‐CLL1‐GISL); GCLLSG V ‐ Rani 2018 (Indian cohort)) (Figure 18).
Rossi (Rossi 2013) (Rossi D ‐ Rossi 2013 (Italian cohort))
We rated the risk of bias of the development study as low across all domains except 'analysis' (Rossi D ‐ Rossi 2013 (Italian cohort)), which we considered to be of unclear risk because handling of missing data handling was not reported. We rated the risk of bias for the two validation studies as unclear due to lack of information on study design for the domain ‘participants’, low for the domains ‘predictors’ and ‘outcome’, and high for the domain ‘analysis’ due to missing data handling and low number of events (Rossi V ‐ Jeromin 2014 (Munich cohort); Rossi V ‐ Rossi 2013 (unclear)) (Figure 21).
We rated the concern for applicability of the development study as low across all domains (Rossi D ‐ Rossi 2013 (Italian cohort)). We rated concern for applicability of the two validation studies as low for the domains 'participants', 'predictors' and 'outcome' (Rossi V ‐ Jeromin 2014 (Munich cohort); Rossi V ‐ Rossi 2013 (unclear)) (Figure 18).
Stephens model (Stephens 2015) (Stephens OS D ‐ Stephens 2015 (Ohio cohort))
We rated the risk of bias for the development study as high for the domains 'participants' (Stephens OS D ‐ Stephens 2015 (Ohio cohort)), because the study included only individuals with del(17p), which implies that only persons with available FISH assessment could have been included. We considered the domains 'predictors' and 'outcomes' to be of low risk. We rated the domain 'analysis' as high due to small sample size, inappropriate missing data handling and inconsistent reporting of performance measures. In a similar manner, we rated the risk of bias for the validation study as high for the domains 'participants' and 'analysis' due to the same reasons, unclear for the domain 'predictors' and low for the domain 'outcome' (Stephens OS V ‐ Stephens 2015 (MDACC)) (Figure 21).
We rated the concern for applicability of the development study (Stephens OS D ‐ Stephens 2015 (Ohio cohort)) and validation study (Stephens OS V ‐ Stephens 2015 (MDACC)) as high for the domain 'participants' due to the selective sample, and low for the domains 'predictors' and 'outcome' (Figure 18).
Prognostic models for the prediction of PFS or TFS (without meta‐analysis, 1 to 3 external validations)
Baliakas model (Baliakas 2019) (Baliakas D ‐ Baliakas 2019 (multicentre))
We rated the risk of bias of the development study as low for the domains ‘predictors’ and ‘outcome’, and high for the domains ‘participants’ since participants with missing data were excluded (Baliakas D ‐ Baliakas 2019 (multicentre)). We rated the domain ‘analysis’ as high due to inappropriate handling of missing data, and univariable selection of predictors. Due to reporting deficiencies concerning inclusion criteria, predictor assessment and outcome definition of the validation study (Baliakas V ‐ Baliakas 2019 (MLL + Scan.)), we rated the risk of bias as unclear across all domains except ‘analysis’, which we considered to be of high risk due to lack of reporting performance measures (Figure 21).
We rated the concern for applicability of the development study as high for the domain ‘participants’ due to the sample not being representative of all CLL patients and low for the domains ‘predictors’ and ‘outcome’ (Baliakas D ‐ Baliakas 2019 (multicentre)). We rated as unclear concern for applicability of the validation study for the domain ‘participants’ due to lack of information about eligibility criteria and recruitment period (Baliakas V ‐ Baliakas 2019 (MLL + Scan.)), and low for the domains ‘predictors’ and ‘outcome’ (Figure 18).
GIMEMA model (Molica 2005) (GIMEMA D ‐ Molica 2005 (GIMEMA cohort))
We rated the risk of bias for the development study as low for the domains ‘participants’ and ‘predictors’ (GIMEMA D ‐ Molica 2005 (GIMEMA cohort)), and high for the domains ‘outcome’ because a predictor was not excluded from the outcome definition and ‘analysis’ due to inappropriate handling of missing data, univariable selection of predictors, and simplification of the model. Likewise, we considered the validation study to have a low risk of bias for the domains ‘participants’ and ‘predictors’ (GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.)), while we rated the domains ‘outcome’ and ‘analysis’ as high risk of bias due to lack of information on performance measures (Figure 21).
We rated the concern for applicability of the development study (GIMEMA D ‐ Molica 2005 (GIMEMA cohort)) and validation study (GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.)) as low across all domains (Figure 18).
MDACC 2011 (Wierda 2011) (MDACC 2011 D ‐ Wierda 2011 (MDACC))
We rated the risk of bias for the development study as high for the domain ‘participants’ due to inappropriate inclusion criteria (MDACC 2011 D ‐ Wierda 2011 (MDACC)), for the ‘outcome’ because outcome definition did not exclude one of the predictors and for ‘analysis’ because of univariable selection of predictors, unclear modelling procedure and lack of performance measures. We only gave a low rating to the domain ‘predictors’. Similarly, we considered the risk of bias of the validation study to be high for the domains ‘outcome’ for the same reason and ‘analysis’ due to unclear handling of missing data and low number of events (MDACC 2011 V ‐ Molica 2016 (O‐CLL1‐GISL)), while we rated the domains ‘predictors’ and ‘participants’ as low risk (Figure 21).
We rated the concern for applicability of the development study as unclear for the domain ‘participants’ due to inappropriate inclusion criteria and low for the domains ‘predictors’ and ‘outcome’ (MDACC 2011 D ‐ Wierda 2011 (MDACC)). We rated concern for applicability of the validation study as low across all domains (MDACC 2011 V ‐ Molica 2016 (O‐CLL1‐GISL)) (Figure 18).
Morabito model (Morabito 2009) (Morabito D ‐ Morabito 2009 (Italian cohort))
We rated the risk of bias of the development study as unclear for the domain ‘participants’ because it was not clear if individuals with missing data were considered (Morabito D ‐ Morabito 2009 (Italian cohort)), low for the domains ‘predictors’ and ‘outcome’, and high for the domain ‘analysis’ due to low number of events, dichotomisation and univariable selection of factors and missing performance measures. In the case of the validation study (Morabito V ‐ Gentile 2014 (O‐CLL1‐GISL)), we rated the risk of bias as low across all domains except ‘analysis’, which we considered to be of unclear risk due to lack of reporting performance measures (Figure 21).
We rated the concern for applicability of the development study as unclear for the domain ‘participants’ due to unclear eligibility criteria, high for the domain ‘predictors’ and low for the domain ‘outcome’ (Morabito D ‐ Morabito 2009 (Italian cohort)). We rated as low, concern for applicability of the validation study across all domains (Morabito V ‐ Gentile 2014 (O‐CLL1‐GISL)) (Figure 18).
O‐CLL1 model (Gentile 2016) (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL))
We rated the risk of bias of the development study as low across the domains ‘participants’ and ‘predictors’ (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL)). We rated the domain 'outcome' as high risk of bias because one predictor was also included in the outcome definition. We rated the domain 'analysis' as high risk of bias due to unclear handling of missing data, categorisation and univariable selection of factors.
Concerning the domain ‘participants’, we rated one validation study as unclear due to missing eligibility criteria (O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)), and one as high risk due to inappropriate inclusion criteria (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort)). We rated both validation studies as low risk of bias for the domain ‘predictors’ and high risk for the domain ‘outcome’ because one predictor was also included in the outcome definition. We considered the risk of bias for the domain ‘analysis’ to be high for one validation study due to the low number of events and no report for handling of missing data (O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)), while we rated the other study as unclear due to unclear handling of missing data (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort)) (Figure 21).
We rated the concern for applicability of the development study as low across all domains (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL)). We rated concern for applicability of the two validation studies (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort); O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)) as unclear for the domains ‘participants’ due to undefined eligibility criteria, the ‘predictors’ as low for the first and unclear for the second due to undefined timing of prognostic factor measurement, and low for both studies for the domain ‘outcome’ (Figure 18).
Stephens model (Stephens 2015) (Stephens TFS D ‐ Stephens 2015 (Ohio cohort))
For risk of bias of the model for TFS in Stephens and colleagues (Stephens TFS D ‐ Stephens 2015 (Ohio cohort)), please see description in the section for OS (above), as the authors used the same development and validation cohorts for both outcomes.
Deficiencies of prognostic model development studies (models with PROBAST rating)
Among the 12 prognostic model development studies with at least one external validation study (including the three meta‐analysed models), we rated nine at high or unclear risk of bias for the domain 'analysis'. We identified the following most common issues in these studies regarding model development.
Missing data handling: only one model development study imputed missing values; all other studies based their analysis on complete‐case analyses.
Predictor selection based on univariable analysis: eight studies derived their multivariable model on prior univariable selection using P values as a criterion.
Correction of estimates: several development studies used split‐sample or bootstrapping techniques. However, none of the studies reported a correction of their regression coefficients to reduce overfitting to the development cohort.
Categorisation of factors: at some stage during the development process, all studies reported categorisation or dichotomisation of continuous prognostic factors.
Model weights: 10 development studies presented a simplification of the original model formula in the form of a point score (weighting, grouping, or counting of disadvantageous prognostic factors), which results in loss of information.
Findings
Reporting of calibration was rare in the studies that we identified. If reported or provided afterwards via email, the format varied so that we were unable to summarise the 'traditional' calibration measures (calibration plots, calibration tables, O:E ratios). Therefore, we decided to use a different way of presenting calibration graphically (Figure 1; Figure 2; Figure 3), based on something that most studies reported (survival frequencies per group). The black lines represent the development study and each coloured line one external validation cohort. The survival per risk group of the development cohort can be interpreted as the expected survival frequencies, and the survival frequencies in the external validation cohorts the observed survival frequencies. The pooled result gives the pooled observed survival frequencies.
19.

Representation of survival per risk group per development and external validation study of the CLL‐IPI (Bahlo 2016)
20.

Representation of survival per risk group per development and external validation study of the Barcelona‐Brno model (Delgado 2017)
21.

Representation of survival per risk group per development and external validation study of the MDACC 2007 model (Wierda 2007)
Models with more than three external validation studies per outcome and meta‐analysis
Prognostic models for the prediction of OS (with meta‐analysis, > 3 external validations)
CLL‐IPI (CLL‐IPI D ‐ Bahlo 2016 (development cohort))
The CLL‐IPI is a prognostic score derived from univariable selection of predictors which were then entered into a forward‐stepwise proportional‐hazards Cox regression model with a hierarchy based on completeness of the predictor information (CLL‐IPI D ‐ Bahlo 2016 (development cohort)). The dataset was split into a training and external validation dataset after univariable selection of factors (66% and 33% of the data, respectively). Continuous factors were dichotomised based on published thresholds and quartiles. The authors integrated prognostic factors that were independently associated with the outcome in the final multivariable model in a weighted manner to construct their prognostic index (Table 2).
| Table 2. Scoring of the CLL‐IPI (Bahlo 2016) | |
| Prognostic factor | Point distribution |
| Age (≥ 65 years) | 1 |
| Clinical stage (Rai I‐IV or Binet B‐C) | 1 |
| IgHV mutational status (unmutated) | 2 |
| B2‐microglobulin (> 3.5 mg/L) | 2 |
| TP53 status (deleted or mutated) | 4 |
| Notes: IgHV: immunoglobulin heavy chain variable region genes | |
Calibration
None of the included studies regarding the CLL‐IPI reported a measure of calibration in their publication.
We included eight external validation studies of this model, with a total of 4891 individuals to calculate the pooled survival frequencies per risk group at five years (Figure 1). The pooled observed survival frequencies of all validation studies were 92.5% (89.2% to 94.8%) for the low‐risk group (score 0‐1), 85.0% (79.7% to 89.1%) for the intermediate‐risk group (score 2‐3), 64.9% (56.4% to 72.6%) for the high‐risk group (score 4‐6) and 40.4% (29.3% to 52.6%) for the very high‐risk group (score 7‐10). For the low, intermediate and high‐risk group, the pooled result of the external validation studies approximated the survival frequencies of the model development study. In the very high‐risk group, survival according to the development study would have been lower than overall in the external validation studies. For the low and intermediate‐risk groups, the 95% PIs were relatively small. The other risk groups showed wide 95% PIs, indicating uncertainty (Table 3), probably due to the low number of individuals in these subgroups (N = 765 and N = 201, respectively). We judged heterogeneity (visual inspection) between external validation cohorts to be low for the calibration of the CLL‐IPI.
| Table 3. Survival per CLL‐IPI risk group | ||||
| Risk group | Risk score | Development study (training dataset): percentage of persons surviving at 5 years (95% CI) | Pooled percentage of persons surviving at 5 years (95% CI) | 95% prediction interval (PI) |
| Low risk | 0‐1 | 93.2% (90.5% to 96.0%) | 92.5% (89.2% to 94.8%) | 82.5% to 97.0% |
| Intermediate risk | 2‐3 | 79.3% (75.5% to 83.2%) | 85.0% (79.7% to 89.1%) | 68.8% to 93.6% |
| High risk | 4‐6 | 63.3% (57.9% to 68.8%) | 64.9% (56.4% to 72.6%) | 41.9% to 82.6% |
| Very high risk | 7‐10 | 23.3% (12.5% to 34.1%) | 40.4% (29.3% to 52.6%) | 13.2% to 72.4% |
Discrimination
We included seven external validation studies with a total of 3307 individuals and 917 deaths observed during the overall observation time. Two studies that presented a measure for the c‐statistic did not report a measure of uncertainty for this measure, which we estimated using the Newcombe method (CLL‐IPI V ‐ Gentile 2016 (Italian cohort); CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort)). For one study (CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)), we included the AUC as a measure of discrimination instead of the c‐statistic. The pooled c‐statistic of the seven studies was 0.72 (95% CI 0.67 to 0.77), with some degree of heterogeneity (between‐study standard deviation, tau = 0.21). The 95% PI ranged from 0.59 to 0.83 (Figure 22).
22.

Meta‐analysis of the c‐statistic for the CLL‐IPI model (Bahlo 2016)
The c‐statistic of the model development study was 0.72 (95% CI 0.68 to 0.75). Based on sensitivity analysis omitting the study that used the AUC instead of the c‐statistic (Figure 23), and comparing estimated and reported 95% CI of the c‐statistic (Figure 24; Figure 25), we conclude that this had no effect on the result. Four out of seven external validation studies replaced the prognostic factor TP53 with a proxy, del(17p); sensitivity analysis showed no substantial differences in discriminative performance (Figure 26).
23.

Sensitivity analysis of the c‐statistic for the CLL‐IPI excluding the study reporting AUC
24.

Sensitivity analysis of the c‐statistic for the CLL‐IPI excluding the study reporting no 95% CI
25.

Sensitivity analysis of the c‐statistic for the CLL‐IPI excluding the study reporting no 95% CI. All 95% CIs were replaced by estimates using the Newcombe method for a comparison with Figure 27
26.

Sensitivity analysis of the c‐statistic for the CLL‐IPI regarding the availability of the predictor TP53 and its proxy del(17p)
Barcelona‐Brno model (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort))
The Barcelona‐Brno score is a prognostic score constructed by comparing the best combinations of the five factors included in another model for CLL (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort)), the CLL‐IPI by Bahlo 2016 CLL‐IPI D ‐ Bahlo 2016 (development cohort)). Incomplete data were most likely part of the exclusion criteria, and therefore persons with missing data were excluded from the analysis in the primary publication. The score classified individuals into three risk groups: persons without del(11q) or del(17p) and mutated IgHV status were classified as low risk; persons with del(11q) or del(17p) and unmutated IgHV status were classified as high risk; all other persons were classified as intermediate risk.
Calibration
None of the included studies in reference to the Barcelona‐Brno model reported a measure of calibration in their publication.
We included three external validation studies of this score with a total of 1974 individuals to calculate the pooled survival frequencies per risk group at five years (Figure 2). The pooled observed survival frequencies of all validation studies were 90.5% (95% CI 85.1% to 94.0%) for the low‐risk group, 79.7% (95% CI 70.7% to 86.5%) for the intermediate‐risk group and 62.5% (95% CI 49.3% to 74.1%) for the high‐risk group. For the low and intermediate‐risk group, the pooled result of the external validation studies approximated the survival frequencies of the model development study. In the high‐risk group, survival according to the development study would have been higher than overall in the external validation studies. For the low‐risk group, the 95% PI was relatively small. However, the intermediate and high‐risk groups showed a wide 95% PI, indicating uncertainty (Table 4). We judged heterogeneity (visual inspection) between external validation cohorts to be low for the calibration of the Barcelona‐Brno score.
| Table 4. Survival per Barcelona‐Brno model risk group | ||||
| Risk group | Risk group | Development study: percentage of persons surviving at 5 years (95% CI) | Pooled percentage of persons surviving at 5 years (95% CI) | 95% prediction interval (PI) |
| Low risk | No del(11q) or del(17p) and mutated IgHV status | 93.4% (90.4% to 96.5%) | 90.5% (85.1% to 94.0%) | 80.4% to 95.7% |
| Intermediate risk | All other | 83.8% (78.4% to 89.6%) | 79.7% (70.7% to 86.5%) | 63.1% to 90.0% |
| High risk | Del(11q) or del(17p) and unmutated IgHV status | 70.0% (59.3% to 82.7%) | 62.5% (49.3% to 74.1%) | 41.3% to 79.7% |
| Notes: IgHV: immunoglobulin heavy chain variable region genes | ||||
Discrimination
For our meta‐analysis of the c‐statistic, we included four external validation studies with a total of 1755 individuals and 416 deaths observed during the overall observation time. Two studies that presented a measure for the c‐statistic did not report a measure of uncertainty, which we estimated using the Newcombe method (Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo); Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.)). The pooled c‐statistic of the four studies was 0.64 (95% CI 0.60 to 0.67), with no heterogeneity between studies (tau = 0.00). The 95% PI ranged from 0.59 to 0.68 (Figure 27).
27.

Meta‐analysis of the c‐statistic for the Barcelona‐Brno model (Delgado 2017)
The c‐statistic of the model development study was 0.68 (95% CI 0.64 to 0.72); it was unclear whether bootstrapping techniques were used to obtain this estimate.
MDACC 2007 (Wierda 2007) (MDACC 2007 D ‐ Wierda 2007 (MDACC))
The MDACC 2007 model is a prognostic model based on univariable selection of predictors that were, when significant, entered into a proportional‐hazards Cox regression model (MDACC 2007 D ‐ Wierda 2007 (MDACC)). Bootstrapping was used to account for optimism in the c‐statistic. The authors provided the precise formula to calculate a point score which could be translated graphically by a nomogram into individual survival probabilities at five or 10 years. In addition, for simplified use in clinical practice, the authors created an index score in a weighted manner (Table 5).
| Table 5. Scoring of the MDACC 2007 model | ||||
| Point distribution | ||||
| Prognostic factor | 0 | 1 | 2 | 3 |
| Age (years) | < 50 | 50‐65 | > 65 | |
| ß‐2 microglobulin (mg/L) | < ULN | 1‐2 × ULN | > 2 × ULN | |
| ALC (× 109/L) | < 20 | 20‐50 | > 50 | |
| Gender | Female | Male | ||
| Rai stage | 0‐II | III‐IV | ||
| No. of involved nodal groups | ≤ 2 | 3 | ||
| Notes: ULN: upper limit of normal; ALC: acute lymphocyte count | ||||
Calibration
The development study provided a calibration plot based on bootstrap sampling. The data points representing the observed versus expected survival probabilities are close to the ideal line. At the higher frequencies, the nomogram slightly underestimates survival. Measures of calibration were not reported in any of the external validation studies.
We included five external validation studies of this model with a total of 3786 individuals for calculating the pooled survival frequencies per risk group at 5 years (graphically represented in Figure 3). The pooled observed survival frequencies of all validation studies was 97.0% (95% CI 94.3% to 98.4%) for the low‐risk group (score 1‐3), 82.3% (74.6% to 88.0%) for the intermediate‐risk group (score 4‐7) and 45.6% (31.3% to 60.5%) for the high‐risk group (score ≥ 8). For the low and intermediate‐risk group, the pooled result of the external validation studies approximated the survival frequencies of the model development study. In the high‐risk group, survival according to the development study would have been higher than overall in the external validation studies. For the low‐risk group, the 95% PI was relatively small. The intermediate and high‐risk group showed wide 95% PIs, indicating uncertainty (Table 6). We judged heterogeneity (visual inspection) of calibration between external validation cohorts to be low for the MDACC 2007 index score.
| Table 6. Survival per MDACC 2007 model risk group | ||||
| Risk group | Index score | Development study: Percentage of persons surviving at 5 years (95% CI) | Pooled percentage of persons surviving at 5 years (95% CI) | 95% prediction interval (PI) |
| Low risk | 1‐3 | 97.0% (95.0% to 99.0%) | 97.0% (94.3% to 98.4%) | 90.9% to 99.0% |
| Intermediate risk | 4‐7 | 80.0% (78.0% to 82.0%) | 82.3% (74.6% to 88.0%) | 61.5% to 93.1% |
| High risk | ≥ 8 | 55.0% (47.2% to 62.8%) | 45.6% (31.3% to 60.5%) | 21.2% to 72.3% |
Discrimination
We included seven external validation studies with a total of 5127 individuals and 994 deaths observed during follow‐up for meta‐analysis of the c‐statistic in the case of the index score. Only one external validation study provided sufficient data on performance for the nomogram, which is presented together with the development study discrimination on the upper part of the forest plot (Figure 28). Four studies that reported a measure for the c‐statistic did not report a measure of uncertainty, which we estimated using the Newcombe method (MDACC 2007 V ‐ Bulian 2011 (Italian‐Swiss); MDACC 2007 V ‐ Gentile 2014 (Italian cohort); MDACC 2007 V ‐ Gentile 2016 (Mayo cohort); MDACC 2007 V ‐ Muñoz‐Novas 2018 (Spanish cohort))). For one study (MDACC 2007 V ‐ González Rodríguez (Cabueñes), we included the AUC as measure of discrimination instead of the c‐statistic. The pooled estimate of the seven studies was 0.65 (95% CI 0.60 to 0.70), with some degree of heterogeneity (between‐study standard deviation, tau = 0.21). The prediction interval, which describes a range for the predicted model discrimination in a new validation study of the model, ranged from 0.51 to 0.77.
28.

Meta‐analysis of the c‐statistic for the MDACC 2007 model (Wierda 2007)
The bootstrap‐corrected c‐statistic of the model development study was 0.84 (95% CI 0.82 to 0.86). The validation study for the nomogram showed a similar discrimination of 0.82 (95% CI 0.79 to 0.85). Other studies that used the nomogram provided the point score of the formula. However, due to the poor graphical quality of the presented nomogram, authors did not attempt to estimate the expected individual survival probabilities per individual (MDACC 2007 V ‐ Bulian 2011 (Italian‐Swiss); MDACC 2007 V ‐ Gentile 2014 (Italian cohort)).
Prognostic models for the prediction of PFS or TFS (with meta‐analysis, > 3 external validations)
We did not identify any prognostic model development studies that were externally validated more than three times for the outcomes PFS, TFS or TFS.
Models with one to three external validation studies
Prognostic models for the prediction of OS (without meta‐analysis, 1 to 3 external validations)
GCLLSG model (Pflug 2014) (GCLLSG D ‐ Pflug 2014 (GCLLSG))
The GCLLSG model was derived from univariable selection of predictors which were entered into a forward and backward stepwise proportional‐hazards Cox regression model (GCLLSG D ‐ Pflug 2014 (GCLLSG)). The authors controlled for several factors specific to the RCT data that were used for model‐building (e.g. study, treatment etc). Bootstrapping techniques were used to test the robustness of the Cox regression model. Persons with missing data were excluded from analysis. Factors were categorised based on published thresholds and quartiles. The authors assigned risk scores to factors, which proved to be independent in the final multivariable model, in a weighted manner to construct the prognostic index presented in Table 7.
| Table 7. Scoring of the GCLLSG model | |
| Prognostic factor | Point distribution |
| FISH category del(17p) | 6 |
| Serum thymidine kinase (> 10.0 U/L) | 2 |
| B2‐microglobulin (> 3.5 mg/L) | 2 |
| B2‐microglobulin (> 1.7 and ≤ 3.5 mg/L) | 1 |
| IgHV mutational status (unmutated) | 1 |
| ECOG PS (> 0) | 1 |
| FISH category del(11q) | 1 |
| Gender (male) | 1 |
| Age (> 60 years) | 1 |
| Notes: FISH: fluorescence in situ hybridisation; IgHV = immunoglobulin heavy chain variable region genes; ECOG PS: Eastern Cooperative Oncology Group Performance Status | |
Four risk groups were determined: low risk (0‐2 points), intermediate risk (3‐5 points), high risk (6‐10 points) and very high risk (> 10 points). Five‐ and six‐year OS was reported for each point score.
Calibration
Calibration was not reported for the development and external validation study (GCLLSG V ‐ Pflug 2014 (Mayo cohort)).
In the development study, survival was 95.2%, 86.9%, 67.6% and 18.7% at five years for the low, intermediate, high and very high‐risk group, respectively. In the validation study, survival was 95.2%, 91.1%, 71.7% and 13.6% at five years for the low, intermediate, high and very high‐risk group, respectively.
Discrimination
The c‐statistic of the prognostic score in the model development study was 0.75 (95% CI 0.70 to 0.78), discrimination in the external validation cohort was 0.77 (95% CI 0.70 to 0.83).
Rossi (Rossi 2013) (Rossi D ‐ Rossi 2013 (Italian cohort))
The prognostic score by Rossi and colleagues was developed in several steps (Rossi D ‐ Rossi 2013 (Italian cohort)). Firstly, the authors identified factors that were independently associated with OS by univariable and multivariable Cox regression analysis. Bootstrapping techniques were used to test the robustness of the model. In the next step, the factors were entered into a decision tree algorithm to divide individuals in subgroups. To test the stability of the decision tree, the random survival forest method with amalgamation algorithm was used. It was unclear how missing data were handled.
Calibration
No study reported a measure of calibration for this model.
In the development study, OS at five years was 86.9% in the very low‐risk group (del13q14 only), 77.6% in the low‐risk group (normal/+12), 65.9% in the intermediate‐risk group (NOTCH1 and/or SF3B1 mutations and/or del11q22‐q23 in the absence of TP53 and BIRC3 abnormalities) and 50.9% in the high‐risk group (TP53 disruption and/or BIRC3 disruption independent of co‐occurring lesions). One external validation study reported median survival per risk group (not reached for the very low‐risk group, 13.8 years for the low‐risk group, 11.2 years for the intermediate‐risk group and 7.7 years for the high‐risk group) (Rossi V ‐ Rossi 2013 (unclear)). In the other validation study, OS at five years was 91% in the very low‐risk group, 90% in the low‐risk group, 75.2% in the intermediate‐risk group and 62.1% in the high‐risk group (Rossi V ‐ Jeromin 2014 (Munich cohort)).
Discrimination
The c‐statistic of the prognostic score in the development cohort was 0.64. Only one of the two validation studies reported a measure of discrimination; the c‐statistic was 0.66 (95% CI not reported).
Stephens model (Stephens 2015) (Stephens OS D ‐ Stephens 2015 (Ohio cohort))
To develop prognostic models for OS, the authors identified significant prognostic factors in multivariable proportional hazards models using backwards selection. Based on the final model, they defined a simplified risk score based on the strength of the association. Missing data were accounted for by multiple imputation techniques. The model and score for OS included the factors ECOG performance status, age and lactate dehydrogenase (LDH).
Calibration
No study reported a measure of calibration for the prognostic models.
In the development cohort, the percentage of alive persons at two years was 89% (95% CI 74% to 96%) for score 0, 66% (95% CI 41% to 82%) for score 2 and 0% for score 4. In the validation cohort, the percentage of alive persons at two years was 95% (95% CI 83% to 99%) for score 0, 80% (95% CI 55% to 92%) for score 2 and 20% (95% CI 1% to 58%) for score 3.
Discrimination
The discrimination of the exact model for OS was 0.76 (P < 0.03) and 0.73 (P < 0.0001) for the development cohort. The validation cohort showed a c‐statistic of 0.68 for the simplified score for OS.
Prognostic models for the prediction of PFS or TFS (without meta‐analysis, 1 to 3 external validations)
Baliakas model (Baliakas 2019) (Baliakas D ‐ Baliakas 2019 (multicentre))
The model presented by Baliakas and colleagues was derived from univariable selection of predictors which were entered into a proportional‐hazards Cox regression model (Baliakas D ‐ Baliakas 2019 (multicentre)), that was internally validated by bootstrapping and further confirmed by recursive partitioning based on conditional inference trees and merging of terminal nodes by an amalgamation algorithm. First, the authors split individuals in two groups, mutated and unmutated IgHV. The risk groups for mutated cases were as follows: (1) low risk: non‐TP53 abnormality/+12/subset #2 membership and Binet stage A; (2) intermediate risk: Binet A with one of the following: TP53 abnormality and/or +12 and/or subset #2 membership, (3) high risk: Binet B, (4) very high risk: Binet C. The risk groups for unmutated cases were as follows: (1) very low risk: non‐TP53abn/SF3B1mut/del(11q) female Binet A; (2) low risk: non‐TP53abn/SF3B1mut/del(11q) male Binet A; (3) intermediate risk: Binet A with one of the following: TP53abn and/or SF3B1mut and/or del(11q); (4) high risk: Binet B; (5) very high risk: Binet C.
Calibration
No measures of calibration were reported for this model.
In the development study, the five‐year treatment probability for mutated cases was 12% for group 1, 40% for group 2, 64% for group 3 and 92% for group 4. For unmutated cases, the five‐year treatment probability was 45% for group 1, 65% for group 2, 78% for group 3, 90% for group 4 and 100% for group 5.
In the external validation cohort, the five‐year treatment probability was only reported for the intermediate‐risk groups (for mutated cases 43%, group 2; and for unmutated cases 74%, group 3).
Discrimination
In the development cohort, the discrimination was 0.745 (SE = 0.013) and 0.753 (SE = 0.013) for the cases with mutated IgHV and unmutated IgHV status, respectively. Discrimination was not reported for the external validation cohort.
GIMEMA model (Molica 2005) (GIMEMA D ‐ Molica 2005 (GIMEMA cohort))
The GIMEMA model was derived from univariable selection of predictors which were entered into a proportional‐hazards Cox regression model (GIMEMA D ‐ Molica 2005 (GIMEMA cohort)). No internal validation was reported. Negative risk factors that were independent in multivariable analysis (lymphocyte doubling time < 12 months, lymphocyte count >30 × 109 per L and Rai stage I to II) were assigned one point each (not weighted for HR) to create a score. Gender was added afterwards as relevant for individuals with score 0, but not for individuals with scores 1 to 3. Three risk groups were created: (1) females with score 0; (2) males with score 0; and (3) individuals with score 1 to 3 of either gender.
Calibration
No measure of calibration was reported for this model.
In the development study, the 10‐year PFS was 76.2% for group 1, 61.4% for group 2 and 37.8% for group 3, respectively. In the validation study, the 10‐year treatment‐free survival was 89% for group 1, 54% for group 2 and 0% for group 3 (GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.)).
Discrimination
The development study did not report a measure of discrimination. The AUC for the validation study was 0.58 (95% CI 0.49 to 0.66).
MDACC 2011 (Wierda 2011) (MDACC 2011 D ‐ Wierda 2011 (MDACC))
The MDACC 2011 model (MDACC 2011 D ‐ Wierda 2011 (MDACC)) is a prognostic model derived from univariable selection of predictors. After univariable selection, the dataset was split into a training and test set. Significant predictors entered into a proportional‐hazards Cox regression model (forward selection procedure with P < 0.1, and removed if not P < 0.5 in final model) were developed on the test set and applied to the test set. The factors of the final model were entered into Cox regression models for 1000 bootstrap samples to show their robustness. The final model formula can be used to derive a point score that can be translated graphically by a nomogram into individual survival probabilities at five or 10 years.
The formula is as follows: [I(No. of lymph node sites involved = 3) × 7.370 + I(FISH11q del) × 9.312 + I(FISH 17p del) × 11.285 + (diameter of largest cervical lymph node in cm) × 4.172 + (LDH/100) × I([IgHV gene mutated] × 5.000 + (LDH/100) × I(IgHV gene = unmutated) × 1.065] + 35.467.
Calibration
No study reported a measure of calibration for this model.
Discrimination
The development study did not report a measure of discrimination. The validation study included a total of 337 participants (91 treated) and reported a c‐statistic of 0.71 (95% CI 0.60 to 0.82) (MDACC 2011 V ‐ Molica 2016 (O‐CLL1‐GISL)).
Morabito model (Morabito 2009) (Morabito D ‐ Morabito 2009 (Italian cohort))
The prognostic model developed by Morabito and colleagues (Morabito D ‐ Morabito 2009 (Italian cohort)) was derived from univariable analysis, including dichotomisation of factors with cut‐off determination by ROC curves. Significant (P > 0.05) prognostic factors were entered into proportional‐hazards Cox regression models. Each unfavourable marker that remained significant was assigned one point, and the sum of these points formed the final score (CD38 positive = 1, ZAP‐70 positive = 1, IgHV unmutated status = 1). Individuals with scores of two or three points were categorised together to form the high‐risk group.
Calibration
No study reported a measure of calibration for this model.
The five‐year PFS of the validation study for this model was 91.7% for the low‐risk group (score 0), 82.9% for the intermediate‐risk group (score 1) and 57.4% for the high‐risk group (score 2‐3) (Morabito V ‐ Gentile 2014 (O‐CLL1‐GISL)).
Discrimination
Both the development and validation study did not report a measure of discrimination.
O‐CLL1 model (Gentile 2016) (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL))
The prognostic score developed by Gentile and colleagues was derived from univariable selection of predictors (O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL)). Before entering the factors into proportional‐hazards Cox regression models (P < 0.05), continuous factors were dichotomised using established thresholds or ROC curves. Bootstrapping techniques were used to test the robustness of the Cox regression model. There were no persons with missing values in the sample. The authors assigned risk scores to each prognostic factor that was significant in multivariable analysis based on their hazard ratio (Table 8). Individuals were divided into three different risk categories for TFS, low (score 0 to 2), intermediate (score 3 to 5) and high risk (score 6 to 7).
| Table 8. Scoring of the O‐CLL1 model | |||
| Point distribution | |||
| Prognostic factor | 0 | 1 | 2 |
| Rai staging system | 0 | I‐II | ‐ |
| B2‐microglobulin | Normal | ‐ | Elevated |
| ALC (109/L) | < 10 | ‐ | ≥ 10 |
| IgHV mutational status | Mutated | ‐ | Unmutated |
| Notes: ALC: acute lymphocyte count; IgHV: immunoglobulin heavy chain variable region genes | |||
Calibration
No study reported a measure of calibration for this model.
In the development study, treatment‐free survival at three years was 95.3% in the low risk, 74.5% in the intermediate‐risk and 28.6% in the high‐risk group. Outcome frequencies were also provided per risk score. One external validation study showed similar percentages of treatment‐free survival at three years with 95.5%, 78.9% and 40.6% for the low, intermediate and high‐risk group, respectively (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort)). The other external validation study presented results in the form of a Kaplan‐Meier curve (O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)).
Discrimination
The c‐statistic of the prognostic score in the model development study was 0.75 (P < 0.001). The c‐statistic in the external validation cohorts were 0.72 (P < 0.001) (O‐CLL1 V ‐ Gentile 2016 (Mayo cohort)), and 0.55 (95% CI 0.49 to 0.60) (O‐CLL‐1 V ‐ Rani 2018 (Indian cohort)).
Stephens model (Stephens 2015) (Stephens TFS D ‐ Stephens 2015 (Ohio cohort))
To develop prognostic models for TFS, the authors identified significant prognostic factors in multivariable proportional hazards models using backwards selection. Based on the final model, they defined a simplified risk score based on the strength of the association. Missing data were accounted for by multiple imputation techniques. The model and score for TFS include the factors ECOG performance status, Rai stage, age, white blood cell count (WBC) and FISH category del(11q22.3). Risk groups were summarised as 0 to 1 points, 2 to 3 points and ≥ 4 points.
Calibration
No study reported a measure of calibration for the prognostic model for TFS.
In the development cohort, the percentage of persons who were treatment‐free at two years was 85% (95% CI 0.60 to 0.95) for the low‐risk group, 51% (95% CI 32 to 67) for the intermediate‐risk group, and 0% for the high‐risk group. In the validation cohort, the percentage of persons who were treatment‐free at three years was 63% (95% CI 39 to 79) for the low‐risk group, 26% (95% CI 15 to 39) for the intermediate‐risk group and 16% (95% CI 6 to 29) for the high‐risk group.
Discrimination
The discrimination of the exact model for TFS was 0.84 (P < 0.017) for the development cohort. The validation cohort showed a c‐statistic of 0.66 for the simplified score for TFS.
Models without any external validation studies
The main characteristics of prognostic models for which no external validation studies could be found are described in Appendix 7.
Subgroup differences
We did not explore subgroup differences as the number of external validation studies per model with available data were too small. Three of the four external validation studies for the Barcelona‐Brno model defined CLL by the International Workshop on CLL Guideline (Hallek 2008), and one did not report diagnostic criteria. Among the seven external validations of the CLL‐IPI, three studies used the International Workshop on CLL Guideline (Hallek 2008), and four did not clearly refer to the diagnostic criteria or used a combination of criteria. One external validation study of the MDACC 2007 model used the International Workshop on CLL Guideline (Hallek 2008), two studies' cohorts were based on the NCI working group criteria (Cheson 1996), and four studies did not clearly report the criteria or used a combination of criteria.
Sensitivity analysis
We planned to explore the effect of risk of bias on the performance measures for the external validation studies per model. We were not able to conduct this analysis as we rated nearly all studies as high or unclear risk of bias, leaving no studies with a low risk of bias for inclusion in the sensitivity analysis.
One external validation study for the CLL‐IPI reported the AUC instead of the c‐statistic (CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)), which we included in our meta‐analysis. The pooled performance estimate for discrimination did not change (0.72, 95% CI 0.67 to 0.77 including the study; 0.72, 95% CI 0.66 to 0.78 excluding the study; see Figure 23). For the MDACC 2007 model, we also included one AUC instead of the c‐statistic in the analysis. By removing this study (MDACC 2007 V ‐ González Rodríguez (Cabueñes)), the pooled performance estimate for discrimination barely changed (from 0.65 to 0.66, 95% CI 0.60 to 0.71; for illustration, see Figure 29).
29.

Sensitivity analysis of the c‐statistic for the MDACC 2007 model excluding the study reporting AUC
To examine the effect of estimating the 95% CIs according to the Newcombe method, we conducted a sensitivity analysis exemplary for one included prognostic model, the CLL‐IPI. In Figure 24, we limited the meta‐analysis of the c‐statistic to all studies that reported the 95% CI, which did not change the overall pooled estimate and 95% CIs substantially (from 0.72, 95% CI 0.67 to 0.77 for the main analysis to 0.73, 95% CI 0.66 to 0.80 for the analysis limited to studies with 95% CI reported). We calculated the 95% CIs based on Newcombe for all studies and the pooled results to compare this extreme scenario with the scenario that all 95% CIs were provided. Figure 25 shows that the estimation did not change the 95% CIs. The 95% PI changed negligibly.
For the CLL‐IPI, one of the predictors was commonly missing (TP53 mutation) and replaced by a proxy prognostic factor (del(17p)). To see if the replacement limited discriminative performance, we conducted separate meta‐analyses (Figure 26). The 95% CI of the pooled c‐statistic overlapped, however, the number of studies in each analysis was very small (N = 3 and N = 4).
Discussion
Summary of main results
We aimed to identify and describe all prognostic models for untreated CLL and their corresponding external validation studies. We identified 12 prognostic models with at least one external validation (Appendix 6). Of these, three models were validated externally more than three times, with appropriate reporting to allow us to conduct a meta‐analysis of the c‐statistic, which is a measure of discriminative performance of a model. Additionally, we identified 40 models or scores without any external validation study, which are further described in Appendix 7 and Characteristics of studies awaiting classification.
Results for models with more than three external validation studies per outcome for meta‐analysis
Prognostic models for the prediction of OS (with meta‐analysis, > 3 external validations)
The pooled c‐statistic of the CLL‐IPI, a point score calculated from five factors, was based on seven external validation studies with a total of 3307 individuals. The pooled estimate of its discriminative performance in seven external validation studies was 0.72 (95% CI 0.67 to 0.77). The 95% PI ranged from 0.59 to 0.83, indicating that from 100 external validation studies for this model, 95 will show a c‐statistic within this range. Calibration was not reported in the publications, and thus the prognostic score was not updated based on calibration. Studies showed somewhat heterogeneous results regarding the pooled survival frequencies (observed survival frequencies) as compared to the development study survival frequencies (expected survival frequencies). Due to our diverging representation of calibration, to make assumptions regarding the calibration of the models, we assumed that the survival frequency of the development cohort can be interpreted as our expected frequency. The pooled survival per risk group was higher than observed in the development study for the very high‐risk group, indicating that the model underpredicted survival in this group. This may be explained by the fact that the model was developed using data including a higher proportion of individuals with treatment indication at recruitment, who are participants with a worse prognosis than the general population with CLL. The curves showed a relatively homogeneous pattern regarding the differences between risk groups, although frequencies varied more widely for the very high‐risk group, probably due to the low number of participants.
The pooled c‐statistic of the Barcelona‐Brno score, which contains two of the prognostic factors also included in the CLL‐IPI, was derived from four external cohorts with a total of 1974 individuals. The pooled c‐statistic was 0.64 (95% CI 0.60 to 0.67) and the 95% PI ranged from 0.59 and 0.68. Results between cohorts were somewhat heterogeneous, although the number of studies was relatively low to draw meaningful conclusions. Calibration was not reported in the publications, and thus the prognostic score was not updated based on calibration. The pooled survival per risk group was lower than observed in the development study, especially for the high‐risk group, which means that across cohorts, the model overestimated survival as compared to the development cohort. This judgement is mainly driven by the Czech cohort. The pattern was consistent across studies.
The pooled c‐statistic of the MDACC 2007 index score, which includes six prognostic factors, was derived from seven external validation studies with a total of 5127 individuals. The pooled c‐statistic was 0.65 (95% CI 0.60 to 0.70; 95% PI: 0.51 to 0.77); discrimination was somewhat heterogeneous between studies. Calibration was not reported in any of the studies validating the index score, thus, no model updates were reported. The pooled survival per risk group matched the survival in the development study for the low and intermediate‐risk groups. In the high‐risk group, the pooled survival per risk group was lower than observed in the development study, thus the model overestimated the survival of the high‐risk group. The pattern was consistent across studies.
From these three prognostic models for untreated individuals with CLL included in meta‐analysis, the CLL‐IPI performed best regarding the discrimination between persons with a 'as good as' compared to a worse prognosis. The score underpredicted survival for the very high‐risk group, which was a small group of individuals defined by TP53 mutation or deletion.
The three models consist of unweighted (Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort)) or weighted (CLL‐IPI D ‐ Bahlo 2016 (development cohort), MDACC 2007 D ‐ Wierda 2007 (MDACC)) simplified point scores, which implies that information concerning the correct weighting of the prognostic factors was lost during simplification. Instead of individual outcome frequencies, the models result in a score on a limited scale, which reduced the possible outcome range compared to the original formula. Consequently, the simplification may have resulted in a reduction of the discriminative performance.
Prognostic models for the prediction of PFS or TFS (with meta‐analysis, > 3 external validations)
We did not identify any prognostic model development studies that were externally validated more than three times for the outcomes PFS, TFS or TFS.
Models with one to three external validation studies
Prognostic models for the prediction of OS (without meta‐analysis, 1 to 3 external validations)
In total, three models were developed to predict OS (GCLLSG D ‐ Pflug 2014 (GCLLSG); Rossi D ‐ Rossi 2013 (Italian cohort); Stephens OS D ‐ Stephens 2015 (Ohio cohort)), with one, two and one external validation study, respectively. Due to the limited information on the predictive performance outside the model development setting, we would not yet consider these models ready for use in clinical practice.
Prognostic models for the prediction of PFS or TFS (without meta‐analysis, 1 to 3 external validations)
In total, one model was developed to predict PFS (GIMEMA D ‐ Molica 2005 (GIMEMA cohort)) and five were developed to predict TFS (Baliakas D ‐ Baliakas 2019 (multicentre); MDACC 2011 D ‐ Wierda 2011 (MDACC); Morabito D ‐ Morabito 2009 (Italian cohort); Stephens TFS D ‐ Stephens 2015 (Ohio cohort); O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL)), with one external validation study per model for the first five and two for the last mentioned. Due to the limited information on the predictive performance outside the model development setting, we would not yet consider these models ready for use in clinical practice.
Models without any external validation studies
An abundance of prognostic models and scores without any application in external cohorts has been published, with a varying degree of reporting quality. Without any further knowledge on their performance in different settings than the development cohort, we would not recommend their use for patients.
Overall completeness, certainty of the evidence and study limitations of externally validated models
Overall completeness of the data
As expected from results of previous research (Heus 2018), reporting of the included models and their validation studies was poor. We have summarised the missing information per model for the development study and external validation studies for the primary model outcome in Appendix 8.
For the CLL‐IPI, none of the studies reported calibration (although we obtained information for five studies from corresponding authors). Reporting of discrimination was higher (8 out of 11, 6 times with 95% CI).
For the Barcelona‐Brno score, none of the studies reported calibration (although we obtained information for one study from the corresponding authors). Reporting of discrimination was slightly higher (5 times out of 6, 3 times with 95% CI).
For the MDACC 2007 index score, two of the studies reported calibration. Reporting of discrimination was slightly higher (8 times out of 10, 7 times with 95% CI).
Of the other nine models with 17 validation studies that were not meta‐analysed, none reported calibration (we obtained information on calibration for one study from the corresponding authors). Reporting of discrimination was higher (12 times, seven times with 95% CI).
Overall, across all categories of included studies (development studies with and without external validations, external validations), information on calibration and discrimination was lacking. Calibration was reported in six studies and discrimination in 37 out of 85 studies.
Many studies either excluded persons with missing prognostic factor data at enrolment, or excluded persons with missing data from the analysis. In most cases, we do not have information on the number of persons in the complete database or sample, and a comparison between included and excluded persons is rarely provided. Instead of removing data of individuals with missing data completely, multiple imputation would be preferable, but should be interpreted carefully (Moons 2019; Steyerberg 2014).
None of the studies explicitly mentioned reasons for censoring, a common phenomenon in time‐to‐event data, in their cohort which may be particularly relevant for studies based on RCT data.
Certainty of the evidence
At the date of submission of this review, no official GRADE guidance for grading the summarised results of meta‐analysis of prognostic models was available. Hence, we refrained from rating the certainty of the evidence.
Study limitations of prognostic model development studies
The majority of PROBAST ratings for model development studies was high. The main reason for this was handling missing data, predictor selection based on univariable selection of predictors, failure to correct estimates for optimism, categorisation of prognostic factors and simplification of models.
Potential bias in the review process
To prevent bias in the review process, we performed all relevant steps in duplicate and solved discrepancies in group discussions. We developed a sensitive search strategy and tracked the references and citations of all included references. However, we did not search conference abstracts or trial registries, since conference abstracts do not provide sufficient information for 'Risk of bias' assessment and prognostic studies are rarely prospectively registered, but rather built on existing retrospective databases. Thus, we would not expect to find any relevant studies by searching those additional databases.
As this is a new review type, our methods evolved during the process. We decided to adapt the newest methods regardless of the protocol, which may have introduced bias.
Prespecification of prognostic factors: we did not limit our inclusion to models with e.g. a minimum set of clinically relevant factors or a specific kind of factor (such as non‐invasive or genetic factors only). Therefore, we made no distinction regarding which predictors were included in the retrieved models – we included all developed and validated models that were the subject of our review, regardless of which type of predictors they included. We did not aim to rate the individual factors, examine the relationship between any one factor with our outcomes or the strength of any one factor above other factors, as this would be the subject of a prognostic factor review.
Search: we have slightly adapted the search to make it more inclusive, and extended it to include studies published since the inception of MEDLINE instead of using the cut‐off year 1990. Based on the recommendation of the Cochrane fast‐track service, we have added a search strategy for Embase. We believe that screening more references has not introduced bias.
Screening: the definition of prognostic models and the reporting standards and, with that, the amount of information expected in a publication has changed substantially over time. To avoid bias, we have included all studies that explicitly stated the aim of developing a score for prognostication, although from today's point of view, we would not consider these scores a well‐developed prediction model, because they did not follow the recommended steps in the model‐building process. Most of these studies can be found in Appendix 7, as models without external validation studies.
Risk of bias: since publication of the protocol, PROBAST was published. In our protocol, we did not specify the data analysis process. To avoid bias and use the most recently developed methods, we worked in close collaboration with the Cochrane Prognosis Methods group, and some authors of PROBAST are co‐authors of this review. For transparency of our group decisions, we added the text under 'Solving disagreements'.
Analysis: we planned to summarise the performance measures for calibration and discrimination. In the review we specified, according to most recent developments, that we would summarise the external validation studies per model. For some studies, we estimated the 95% CI for the c‐statistic due to lack of reporting. Based on our sensitivity analysis, we assume that this estimation has not introduced bias.
We did not assess the likelihood of publication bias, i.e. a preferential publication of studies which show better model performance, because currently, there is no established standard for this.
We would like to mention that external validation studies are ideally carried out by independent researchers (using independent samples). For the studies included in this review, this was not always the case (e.g. CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014); CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort)), and may have led to additional bias to the one assessed by PROBAST.
Applicability of findings to clinical practice and policy
We identified 52 prognostic models for various outcomes. Due to the lack of external validation, we disregarded 40 of these as not (yet) applicable. Nine more prognostic models were validated only one to three times, which we would not consider to be sufficient information to make a judgement of applicability to different settings. The three models included in meta‐analysis of the c‐statistic offer more information concerning their performance in different populations. The CLL‐IPI seems to perform best among the three models in both the development and external validation cohorts. However, due to the relatively small number of external validation studies (N < 10) and serious limitations in reporting, we cannot draw final conclusions regarding the generalisability and usefulness of this model in clinical practice.
The three models that were included in meta‐analyses all predict OS. Until 2015, treatment options for individuals with CLL had not changed drastically over time. However, since the introduction of new agents, such as ibrutinib and idelalisib, survival has improved for all risk groups. For individuals with a poor prognosis, there is hope that they will benefit from new and better tailored treatments. Improvements in survival for all risk groups results in systematic underprediction of survival of prognostic models that were developed using data of patients receiving traditional treatment regimens, while the improvement of prognosis especially for high‐risk individuals will lead to lowered discriminative ability of a prognostic model. The prognostic models discussed in detail in this review may thus need to be updated, taking this improved baseline risk into account. In contrast, other patient‐relevant outcomes, such as time‐to‐treatment or progression would not be influenced by improved treatment options as long as a watch‐and‐wait strategy is adopted, and may be an interesting alternative outcome for prediction models. Some of the models were already externally validated for both of these outcomes, and may be included in a future update of this review.
A further limitation of prognostic models in general is that they usually consider only one outcome per model, thereby not considering a balance between different outcomes, such as survival, quality of life, and other outcomes of interest.
CLL has experienced several changes in diagnostic criteria over time (in particular, in 2008 the criterion of having an absolute lymphocyte count (ALC) more than or equal to 5.0 × 109/L2 to a B‐cell count more than or equal to 5.0 × 109/L), changing the categorisation of individuals with clinical monoclonal lymphocytosis (cMBL) or CLL (Cheson 1996; Hallek 2008). We aimed to explore this change in subgroup analysis but did not, due to the low number of included studies with clear division. However, this change is not expected to have an extensive effect on the performance of a model, as prognosis and patient characteristics are comparable in individuals with cMBL and Rai stage I.
Agreements and disagreements with other studies or reviews
To our knowledge, the current review is the first systematic review that aimed to identify all prognostic models developed for untreated individuals with CLL and their corresponding external validation studies.
We are aware of two other systematic reviews that cover one of the prognostic models that we identified, the CLL‐IPI (Molica 2018a; Molica 2018b). These systematic reviews aimed at identifying all published studies that have used CLL‐IPI to predict the clinical outcome of CLL; one in patients who received chemoimmunotherapy or targeted therapies (Molica 2018a), the other without restriction of therapy (Molica 2018b). There are some differences between these reviews and our systematic review. In Molica 2018a, studies were limited to individuals who received chemoimmunotherapy or targeted therapies and also included studies of relapsed/refractory CLL from conference abstracts. The authors summarised the survival frequencies per CLL‐IPI risk group at two years for OS, whereas we pooled OS at five years. Molica 2018b included patients undergoing any treatment, and summarised both OS and TFS at five years. Both these reviews included data from conference proceedings, whereas we did not. Moreover, they did not report measures of discrimination such as the c‐statistic (which is pivotal when assessing model performance) and did not assess risk of bias or applicability.
The included studies in Molica 2018b mostly overlapped with our identified studies for OS. The exception was two external validation studies published as abstracts (N = 2, CLL11 study population; Goede 2016 and Ferrer Lores 2016). In contrast, we identified three additional cohorts which had not yet been published at the time of publication of Molica 2018a and Molica 2018b (CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort); CLL‐IPI V ‐ Rani 2018 (Indian cohort); CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)).
Authors' conclusions
Implications for practice
We identified three prognostic models that were validated more than three times for their primary outcome OS: the CLL‐IPI, the Barcelona‐Brno score, and the MDACC 2007 index score. Of the three models, the CLL‐IPI seems to perform best regarding the discrimination between individuals with a good prognosis as compared to individuals with a worse prognosis. However, the number of external validation studies was relatively low to draw definite conclusions. Especially for the very high‐risk group, the index underpredicts survival, which may be a result of the selective sample that was used to develop the index.
This review has been developed in a time of rapidly expanding treatment options that may drastically change the prognostic implications for the therapy and longevity of these patients. The external validation cohorts may not be representative anymore of the currently available, improved treatment options. Therefore, the models can be used to provide an approximate classification, but may need testing and updating before being applied to new patients.
Implications for research
This systematic review shows that for CLL, an abundance of studies of prognostic models and scores can be identified. Based on our very inclusive definition of a prognostic score, we found that these studies (including the newer studies) were not performed according to the current standards of prognostic model development. For the future, we recommend authors of model development studies incorporate the most recent checklists and tools, such as CHARMS and TRIPOD, as orientation to avoid common issues. We would also like to emphasise that reporting in more recent papers can benefit from improvements. The minimum reported information should include participant information (recruitment, study design, eligibility criteria, diagnostic criteria used, etc.), predictor assessment, outcome definition, and the relevant performance measures.
Our review also shows that the proportion of external model validation studies as compared to model development studies is very low. Before investing in the development of a new model, we recommend the validation and direct comparison of the existing models in different settings and populations. Most suitable for this aim are well‐designed prospective studies that also take into account novel agents and emerging molecular makers. To improve predictive performance, a model can be updated with these markers or tailored to a specific population.
With time, newly identified prognostic factors may be added.
History
Protocol first published: Issue 1, 2016 Review first published: Issue 7, 2020
Notes
Parts of this review, especially the methods, are from the Cochrane Haematology standard template.
Acknowledgements
The research was supported by National Health Service (NHS) Blood and Transplant and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC). The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health.
We would like to thank Sarah Hodgkinson (Associate Editor, Cochrane Editorial and Methods Department), Helen Wakeford and Clare Dooley (Managing Editors, Cochrane Editorial and Methods Department), and Anne Lethaby (Copy Editor, Copy Edit Support) for their assistance regarding the review. We would also like to thank Robin Featherstone (Information Specialist, Cochrane Editorial and Methods Department) for suggestions to improve our search strategy.
We also thank Alexandra McAleenan (Methods), Lucinda Archer (Methods), Sherry Reisner (Health Writer), Dr Stefano Molica (Department of Hematology‐Oncology, Azienda Ospedaliera Pugliese‐Ciaccio, Catanzaro, Italy), Dr Mary Ann Anderson (Clinical Haematology Department of The Royal Melbourne Hospital and Peter MacCallum Cancer Centre; Blood Cells and Blood Cancer Division of the Walter and Eliza Hall Institute, Melbourne, Australia) and Dr SG Agrawal (Senior Lecturer and Honorary Consultant, Division of Haemato‐Oncology, St Bartholomew's Hospital, Barts Health NHS Trust; and Centre for Immunobiology, Blizard Institute, Queen Mary, University of London) for their valuable peer review.
We would like to thank Thomas Debray from the Cochrane Prognosis Methods group for statistical advice and Natali Pflug for clinical advice.
We would like to thank Yuan Chi (Centre for Evidence‐Based Chinese Medicine, Beijing University of Chinese Medicine) for the translation of a Chinese publication (CLL‐IPI V ‐ Zhu 2018 (Chinese cohort)) and Leonardo Perales‐Guerrero (medical student, Universidad de Guadalajara, Mexico) for the translation of a Spanish publication (GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.)).
We thank all principal investigators who have replied to our inquiries and/or provided us with further information or data concerning their publications. Their help enabled us to include as many studies as possible in analysis. We want to thank Massimo Gentile, Caspar da Cunha‐Bang, Carolina Muñoz‐Novas, Lata Rani, Gian Matteo Rigolin, Huayuan Zhu, Julio Delgado, Sanja Trajkova, Natali Pflug, Jasmin Bahlo, Pietro Bulian, and Neil E Kay.
Appendices
Appendix 1. MEDLINE (via Ovid) search strategy up to 19 September 2018
| 1 | exp LEUKEMIA, LYMPHOCYTIC, CHRONIC, B‐CELL/ |
| 2 | ((lymphocytic* or b‐cell or b‐lymphocytic or lymphoblastic or lymphatic) adj1 leuk?em$ adj3 (chronic$ or cronic$ or chroniq$ or well‐differentia$)).tw,kf,ot. |
| 3 | ((lymphoplasmacytoid or lymphocytic) adj1 lymphom* adj3 (chronic$ or cronic$ or chroniq$ or well‐differentia$)).tw,kf,ot. |
| 4 | ((small cell$ or small‐cell$) adj3 lymphom$).tw,kf,ot. |
| 5 | (lymphom$ adj2 lymphocyt$).tw,kf,ot. |
| 6 | lymphoplasma?ytoid.tw,kf,ot. |
| 7 | (cll or b‐cll or bcll).tw. |
| 8 | sll.tw. |
| 9 | or/1‐8 |
| 10 | (predict$ or clinical$ or outcome$ or risk$).mp. |
| 11 | validat$.mp. or predict$.ti. or rule$.mp. |
| 12 | (predict$ and (outcome$ or risk$ or models$)).mp. |
| 13 | ((history or variable$ or criteria or scor$ or characteristic$ or finding$ or factor$) and (predict$ or model$ or decision$ or identif$ or prognos$)).mp. |
| 14 | decision$.mp. and ((model$ or clinical$).mp. or LOGISTIC MODELS/) |
| 15 | (prognostic and (history or variable$ or criteria$ or scor$ or characteristic$ or finding$ or factor$ or model$)).mp. |
| 16 | or/10‐15 |
| 17 | validat$.mp. or predict$.ti. or DECISION SUPPORT TECHNIQUES/ or rule$.mp. or PREDICTIVE VALUE OF TESTS/ |
| 18 | (predict$ and (clinical$ or identif$)).mp. |
| 19 | 17 or 18 |
| 20 | RISK ASSESSMENT/ |
| 21 | (risk$ adj scores$).tw,kf,ot. |
| 22 | exp RISK FACTORS/ |
| 23 | (risk$ adj (score$ and factor$)).tw,kf,ot. |
| 24 | DECISION SUPPORT TECHNIQUES/ |
| 25 | (decision$ adj2 (techniqu$ or model$)).tw,kf,ot. |
| 26 | (decision$ and support$ and technique$).tw,kf,ot. |
| 27 | (prediction$ and rule$ and clinical$).tw,kf,ot. |
| 28 | (decision$ adj2 (modeling$ or aid$ or analys$ or technique$)).tw,kf,ot. |
| 29 | or/20‐28 |
| 30 | ANIMALS/ not HUMANS/ |
| 31 | 9 and (16 or 19 or 29) |
| 32 | 31 not 30 |
Appendix 2. MEDLINE (via Ovid) search strategy, 19 September 2018 to 24 June 2019, including study filter
| 1 | exp LEUKEMIA, LYMPHOCYTIC, CHRONIC, B‐CELL/ | ||
| 2 | ((lymphocytic* or b‐cell or b‐lymphocytic or lymphoblastic or lymphatic) adj1 leuk?em$ adj3 (chronic$ or cronic$ or chroniq$ or well‐differentia$)).tw,kf,ot. | ||
| 3 | ((lymphoplasmacytoid or lymphocytic) adj1 lymphom* adj3 (chronic$ or cronic$ or chroniq$ or well‐differentia$)).tw,kf,ot. | ||
| 4 | ((small cell$ or small‐cell$) adj3 lymphom$).tw,kf,ot. | ||
| 5 | (lymphom$ adj2 lymphocyt$).tw,kf,ot. | ||
| 6 | lymphoplasma?ytoid.tw,kf,ot. | ||
| 7 | (cll or b‐cll or bcll).tw. | ||
| 8 | sll.tw. | ||
| 9 | or/1‐8 | ||
| 10 | Validat$.tw. or Predict$.ti. or Rule$.tw. or (Predict$ and (Outcome$ or Risk$ or Model$)).tw. or ((History or Variable$ or Criteria or Scor$ or Characteristic$ or Finding$ or Factor$) and (Predict$ or Model$ or Decision$ or Identif$ or Prognos$)).tw. or (Decision$.tw. and ((Model$ or Clinical$).tw. or LOGISTIC MODELS/)) or (Prognostic and (History or Variable$ or Criteria or Scor$ or Characteristic$ or Finding$ or Factor$ or Model$)).tw. or ("Stratification" or "Discrimination" or "Discriminate" or c‐statistic or "Area under the curve" or "AUC" or "Calibration" or "Indices" or "Algorithm" or "Multivariable").tw. | ||
| 11 | 9 and 10 | ||
| 12 | exp ANIMALS/ not HUMANS/ | ||
| 13 | 11 not 12 | ||
| 14 | limit 13 to ed=20180919‐20190624 |
key: exp # /: explode # MeSH subject heading, tw: text word, kf: keyword heading word, ot: original title, * or $: truncation, ?: wildcard, adj#: adjacent within # number of words search line #10: Geersing G‐J, Bouwmeester W, Zuithoff P, Spijker R, Leeflang M, Moons KG, et al. (2012) Search filters for finding prognostic and diagnostic prediction studies in Medline to enhance systematic reviews. PLOS One 2012; 7(2): e32844, doi:10.1371/journal.pone.0032844
Appendix 3. Embase search strategy up to 24 June 2019
| 1 | exp CHRONIC LYMPHATIC LEUKEMIA/ |
| 2 | ((lymphocytic* or b‐cell or b‐lymphocytic or lymphoblastic or lymphatic) adj1 leuk?em* adj3 (chronic* or cronic* or chroniq* or well‐differentia*)).tw,kw. |
| 3 | ((lymphoplasmacytoid or lymphocytic) adj1 lymphom* adj3 (chronic* or cronic* or chroniq* or well‐differentia*)).tw,kw. |
| 4 | ((small cell* or small‐cell*) adj3 lymphom*).tw,kw. |
| 5 | (lymphoma* adj2 lymphocyt*).tw,kw. |
| 6 | lymphoplasma?ytoid.tw,kw. |
| 7 | (cll or b‐cll or bcll).tw. |
| 8 | sll.tw. |
| 9 | or/1‐8 |
| 10 | Validat*.tw. or Predict*.ti. or Rule*.tw. or (Predict* and (Outcome* or Risk* or Model*)).tw. or ((History or Variable* or Criteria or Scor* or Characteristic* or Finding* or Factor*) and (Predict* or Model* or Decision* or Identif* or Prognos*)).tw. or (Decision*.tw. and ((Model* or Clinical*).tw. or STATISTICAL MODEL/)) or (Prognostic and (History or Variable* or Criteria or Scor* or Characteristic* or Finding* or Factor* or Model*)).tw. or ("Stratification" or "Discrimination" or "Discriminate" or c‐statistic or "Area under the curve" or "AUC" or "Calibration" or "Indices" or "Algorithm" or "Multivariable").tw. |
| 11 | 9 and 10 |
| 12 | exp ANIMAL/ not HUMAN/ |
| 13 | 11 not 12 |
| 14 | limit 13 to embase |
| 15 | limit 14 to em=197401‐201926 |
key: exp # /: explode # MeSH subject heading, tw: text word, kw: keyword, *: truncation, ?: wildcard, adj#: adjacent within # number of words searchline #10: Geersing G‐J, Bouwmeester W, Zuithoff P, Spijker R, Leeflang M, Moons KG, et al. Search filters for finding prognostic and diagnostic prediction studies in Medline to enhance systematic reviews. PLOS One 2012; 7(2): e32844, doi:10.1371/journal.pone.0032844 (translated in Embase)
Appendix 4. ClinicalTrials.gov search strategy up to 05 March 2020
Basic search
prognostic OR predictive OR model OR score | chronic lymphocytic leukemia OR CLL
Advanced search
Intervention/treatment: prognostic OR predictive OR Model OR score
Recruitment: all studies
Study type: all studies
Appendix 5. WHO ICTRP search strategy
Basic search
Chronic lymphocytic leukemia AND prognostic
Chronic lymphocytic leukemia AND predictive
Chronic lymphocytic leukemia AND model
Chronic lymphocytic leukemia AND score
CLL AND prognostic
CLL AND predictive
CLL AND model
CLL AND score
Advanced search
Condition: chronic lymphocytic leukemia OR CLL
Intervention: prognostic OR predictive Or model OR score
Recruitment status: ALL
Appendix 6. Prognostic models with external validation studies
| Author, year | Model (= 1) or Score (= 2) | Outcome | Recruitment period | Number of patients | Study design | Number of predictors included in final model | Discrimination | Calibration |
| Delgado 2017 | 2 | OS | NR | 524 | Retrospective cohort | IgHV; FISH abnormalities (2) |
0.682 | Calibration plot for survival at 5 years |
| International CLL IPI working group | 2 | OS | 1997 to 2007 | 3472 | RCTs | IgHV; B2M; age; stage; TP53 (5) |
0.723 | NR |
| Morabito 2009 | 2 | OS | NR | 262 | Unclear | IgHV; ZAP‐70; CD38 (3) |
NR | NR |
| Pflug 2014 | 2 | OS | 1997 to 2006 | 1223 | RCTs | IgHV; B2M; age; gender; del17p; ECOG; sTK; del11q (8) |
0.75 | NR |
| Wierda 2007 | 1 | OS | 1981 to 2004 | 1674 | Retrospective cohort | IgHV; B2M; age; stage; gender; ALC (6) |
0.84 | Calibration plot for survival at 5 years |
| Stephens 2015 | 2 | OS | 2002 to 2012 | 114 | Retrospective cohort | age; LDH; ECOG (3) |
0.73 | NR |
| Molica 2005 | 2 | PFS | 1991 to 2000 | 1138 | Retrospective cohort | stage; gender; lymphocytosis; LDT (4) |
NR | NR |
| Baliakas 2019 | 2 | TFS | NR | 1900 | Retrospective cohort | IgHV; TP53; FISH abnormalities; gender (4) |
0.745 | NR |
| Gentile 2016 | 2 | TFS | 2007 to NR | 480 | Prospective cohort | IgHV; B2M; stage; ALC (4) |
0.75 | Hosmer‐May test |
| Rossi 2013 | 2 | TFS | NR | 673 | Retrospective cohort | TP53; FISH abnormalities; SF3B1 (3) |
0.642 | Bias‐corrected calibration slope, 0.965 |
| Wierda 2011 | 1 | TFS | 2004 to 2009 | 930 | Retrospective cohort | IgHV; LDH; number of involved lymph nodes; FISH abnormalities; lymph node size in neck; interaction term (6) |
NR | NR |
| Stephens 2015 | 2 | TFS | 2002 to 2012 | 114 | Retrospective cohort | age; stage; ECOG; del11q; WBC count (5) |
0.84 | NR |
| Abbreviations: OS: overall survival, PFS: progression‐free survival; TFS: treatment‐free survival; NR: not reported; RCT: randomised controlled trial; IgHV: immunoglobulin heavy chain variable region gene mutational status; B2M: beta 2 microglobulin; FISH: fluorescence in situ hybridisation genomic aberrations; Zap‐70: zeta‐chain‐associated protein kinase 70; CD38: cluster of differentiation 38; LDH: lactate dehydrogenase; LPL: lipoprotein lipase; ECOG: Eastern Cooperative Oncology Group performance status | ||||||||
Appendix 7. Prognostic models without external validation studies
| Author, year | Model (=1) or Score (=2) | Outcome | Recruitment period | Number of patients | Study design | Predictors included in final model (number) | Discrimination | Calibration |
| Baumann 2014 | 2 | OS | 1990 to 2012 | 949 | Retrospective cohort | B2M; stage; Zap‐70; Comorbirities (4) |
NR | NR |
| Bulian 2012 | 1 | OS | 1996 to 2008 | 620 | Retrospective cohort | IgHV; B2M; age stage; FISH; gender (6) |
0.79 | NR |
| Bulian 2011 | 1 | OS | 1996 to NR | 1480 | Retrospective cohort | B2M; age; stage; gender (4) |
0.78 | Calibration plot for survival at 5 years |
| Furundarena 1994 | 1 | OS | 1973 to 1992 | 150 | Retrospective cohort | age; stage; gender; splenomegaly (4) |
NR | NR |
| Haferlach 2010 | 2 | OS | 2005 to 2008 | 399 | NR | IgHV; age; TP53; translocation IGH@ on 14q32; number of chromosome aberrations based on CBA; WBC count (6) |
NR | NR |
| Jarque 1991, model A | 1 | OS | 1969 to 1988 | 187 | Retrospective cohort | Age; Spinal infiltration; BUN (3) |
NR | NR |
| Jarque 1991, model B | 1 | OS | 1969 to 1988 | 187 | Retrospective cohort | Albumin; Spinal infiltration; BUN; cervical adenopathies (4) |
NR | NR |
| Lee 1987 | 1, 2 | OS | 1970 to 1983 | 325 | Retrospective cohort | Age; LDH; uric acid; alkaline phosphatase; external lymphadenopathy (5) |
NR | NR |
| Liang 2018, model 3a | 2 | OS | 2000 to 2014 | 501 | Retrospective cohort | IgHV; B2M; stage; TP53; albumin; HBV infection (6) |
time‐dependent ROC curve | NR |
| Molica 1990 | 1 | OS | NR | 221 | Unclear | age; stage; LAR (3) |
NR | O/E ratios |
| Molica 2019 | 1, 2 | OS | 2000 to NR | 108 | Retrospective cohort | age; ADL; CIRS (3) |
c = 0.70 (95% CI 0.53 to 0.87) | NR |
| Pepper 2012 | 1 | OS | NR | 1154 | Unclear | IgHV; age; CD38; LDT (4) |
NR | NR |
| Rozman 1982 | 1 | OS | NR | 150 | Unclear | Splenomegaly; lymphocytosis; anaemia; thrombocytopenia (4) |
NR | O/E probabilities |
| Rozman 1984 | 1 | OS | NR | 329 | Unclear | lymphadenopathy; haemoglobin; bone marrow pattern; hepatomegaly (4) |
NR | O/E ratios |
| Stamatopoulos 2010 | 2 | OS | NR | 170 | Unclear | ZAP‐70; LPL; miR‐29 (3) |
NR | NR |
| Tsimberidou 2007 | 2 | OS | 1985 to 2005 | 1893 | Retrospective cohort | Age; B2M; Del17p; albumin; creatinine (5) |
NR | NR |
| Visentin 2015 | 2 | OS | 1983 to 2013 | 608 | Retrospective cohort | IgHV; CD38; FISH abnormalities (3) |
0.88 | NR |
| Wierda 2009 | 1, 2 | OS | 1985 to 2004 | 595 | Unclear | Age; B2M; treatment (3) |
NR | Concordance index for calibration curve (nomogram): 0.81 |
| Friedrichs 2011 | 2 | PFS | NR | 134 | NR | IgHV; CD38; iLR (3) |
NR | NR |
| Leotard 2000 | 1 | PFS | 1985 to 1997 | 88 | Prospective cohort | B2M; LDH; albumin; sCD23 (4) |
NR | NR |
| Letestu 2010 | 1, 2 | PFS | NR | 339 | NR | B2M; CD38; lymphocytosis; sTK (4) |
NR | NR |
| Antic 2011 | 2 | TFS | NR | 33 | Unclear | B2M; LPL; sVEGF (3) |
NR | NR |
| Cavallini 2017 | 2 | TFS | NR | 125 | NR | SF3B1; ERK1/2 phosphorylation (2) |
NR | NR |
| Del Guidice 2005 | 2 | TFS | 2003 to 2004 | 201 | Prospective cohort | ZAP‐70; CD38 (2) |
NR | NR |
| Gentile 2009 | 2 | TFS | NR | 222 | NR | IgHV; B2M; CD38 (3) |
NR | NR |
| Haferlach 2010 | 2 | TTT | 2005 to 2008 | 399 | NR | IgHV; ATM deletion; translocation involving IGH@ locus on 14q32; number of chromosome abnormalities (4) |
NR | NR |
| Li 2017b | 2 | TFS | 2007 to 2015 | 406 | NR | IgHV; stage; Del17p (3) |
NR | NR |
| Liang 2018 model 3b | 2 | TFS | 2000 to 2014 | 501 | Retrospective cohort | IgHV; LD; lymphocytosis; platelets; HBV (5) |
time‐dependent ROC curve | NR |
| Metze 2000 | 2 | TFS | 1995 to 1998 | 57 | Prospective cohort | total tumor mass score; percentage of cells with 1 AgNOR cluster (2) |
NR | NR |
|
Miao 2019, model A (CLL‐IPI‐S) |
1, 2 | TFS | 2000 to 2017 | 399 | Retrospective cohort | IgHV; B2M; Age; stage; TP53; SF3B1 (6) |
AUC = 0.762 | calibration plot |
|
Miao 2019, model B (CLL‐PI) |
1, 2 | TFS | 2000 to 2017 | 399 | Retrospective cohort | IgHV; B2M; stage; TP53; SF3B1 (5) |
AUC = 0.773 | calibration plot |
| Molica 2010 | 2 | TFS | 1998 to 2008 | 150 | Retrospective cohort | IgHV; BAFF (2) |
0.86 | NR |
| Molica 2015 | 1 | TFS | NR | 322 | Unclear | IgHV; B‐cell count; interaction term (3) |
NR | NR |
| Morabito 2011 | 2 | TFS | NR | 449 | Retrospective cohort | stage; ZAP‐70; FISH abnormalities; SFLC (kappa + delta) (4) |
NR | NR |
| Pepper 2012 | 1 | TFS | NR | 1154 | Unclear | IgHV; age; LDT; CD38 (4) |
NR | NR |
| Qin 2018 | 2 | TTT | 2008 to 2016 | 334 | Retrospective cohort | IgHV; EBV DNA positive; stage; TP53; ALC; HBsAg+; (6) |
AUC = 0.768 | NR |
| Schweighofer 2011 | 1 | TFS | NR | 131 | Retrospective cohort | SKI (gene); SLAMF1 (gene) (2) |
NR | NR |
| Stamatopoulos 2010 | 2 | TFS | NR | 170 | Unclear | ZAP‐70; LPL; miR‐29c (3) |
NR | NR |
| Vetro 2018 | 1 | TTT | NR | 171 | Unclear | IgHV; SF3B1; percentage of B cells; genomic aberrations (4) |
NR | NR |
| Wierda 2009 | 1 | TFS | 1985 to 2004 | 595 | Unclear | Age; B2M; treatment; bone marrow lymphocytes (4) |
NR | NR |
| Abbreviations: OS: overall survival, PFS: progression‐free survival; TFS: treatment‐free survival; TTT: time to treatment; NR: not reported; RCT: randomised controlled trial; IgHV: immunoglobulin heavy chain variable region gene mutational status; B2M: beta 2 microglobulin; FISH: fluorescence in situ hybridisation genomic aberrations; Zap‐70: zeta‐chain‐associated protein kinase 70; CD38: cluster of differentiation 38; LDH: lactate dehydrogenase; LPL: lipoprotein lipase; ECOG: Eastern Cooperative Oncology Group performance status; LAR: lymphocyte accumulation time; sVEGF: soluble vascular endothelial growth factor; iLR: immature laminin receptor; CIRS: Cumulative Illness Rating Score; ADL: Katz Activity of Daily Living; EBV: Epstein‐Barr virus; HBsAg: surface antigen of hepatitis B virus | ||||||||
Appendix 8. Reporting deficiencies in all included studies with at least one external validation study (development + validation studies, listed per model)
| Prognostic model | Number of cohorts | Recruitment period reported in: | Study design reported in: | Observation time (range) reported in: | Total sample size reported in: | Number of events reported in: | Calibration reported in: | Discrimination (95% CI or SE) reported in: |
| Baliakas (Baliakas 2019) | 2 | 0 | 2 | 1 (1) | 2 | 0 | 0 | 1 (1) |
|
Barcelona‐Brno (Delga do 2017) |
6 | 3 | 4 | 6 (6) | 6 | 5 | 1 | 5 (3) |
| CLL‐IPI (Bahlo 2016) | 11 | 9 | 9 | 10 (6) | 10 | 9 | 5 | 8 (6) |
| GCLLSG (Pflug 2014) | 2 | 1 | 2 | 2 (0) | 2 | 2 | 2 | 2 (2) |
| GIMEMA (Molica 2005) | 2 | 2 | 2 | 1 (1) | 2 | 1 | 0 | 1 (1) |
| MDACC 2007 (Wierda 2007) | 10 | 7 | 8 | 8 (9) | 9 | 9 | 2 | 8 (7) |
| MDACC 2011 (Wierda 2011) | 2 | 1 | 2 | 2 (2) | 2 | 2 | 0 | 1 (1) |
| Morabito model (Morabito, 2009) | 2 | 2 | 1 | 2 (2) | 2 | 2 | 0 | 0 (0) |
| O‐CLL1 model (Gentile 2016) | 3 | 1 | 1 | 3 (2) | 3 | 3 | 0 | 3 (2) |
| Rossi (Rossi 2013) | 3 | 3 | 3 | 3 (0) | 3 | 2 | 0 | 2 (0) |
| Stephens (Stephens 2015) | 2 | 1 | 1 | 0 (0) | 2 | 0 | 0 | 2 (0) |
| Stephens (Stephens 2015) | 2 | 1 | 1 | 0 (0) | 2 | 0 | 0 | 2 (0) |
Characteristics of studies
Characteristics of included studies [ordered by study ID]
Baliakas D ‐ Baliakas 2019 (multicentre).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants and number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding and conflict of interest
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Missing data used as a reason for exclusion: only participants: (quote) "for whom immunogenetic data was available were included in this multicentre retrospective study". |
| Domain 2: Predictors | Yes | Detailed description of predictor assessment in the appendix |
| Domain 3: Outcome | Yes | Predefined outcome definition |
| Domain 4: Analysis | No | Participants with missing data were excluded from multivariable analyses: (quote) "we considered only those cases with available data for all the factors included in the model (n = 918 for M‐CLL and n = 384 for U‐CLL)"; univariable selection of predictors; assumptions of Cox proportional hazards model were checked. |
| Overall judgement | No | |
Baliakas V ‐ Baliakas 2019 (MLL + Scan.).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants and number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding and conflict of interest
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Eligibility criteria not reported; not stated whether participants were excluded based on missing values; recruitment period not reported |
| Domain 2: Predictors | Unclear | Lack of information about predictor assessment, especially since the validation cohort consisted of two separate cohorts which were merged for analysis |
| Domain 3: Outcome | Unclear | Outcome definition not reported |
| Domain 4: Analysis | No | Performance measures (calibration or discrimination) not reported |
| Overall judgement | No | |
Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants and number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding and conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Retrospective retrieval of individuals with available information for their model ‐ individuals without the necessary data may have been left out. Unclear recruitment period |
| Domain 2: Predictors | Yes | No explicit statement on predictor measurement, however, there are indications that in this single‐centre study, lab procedures remained similar. This was a retrospective cohort, possibility to look forward |
| Domain 3: Outcome | Yes | No clear description of outcome assessment. We assumed that assessment of the objective outcome OS was similar within this single‐centre study and therefore did not rate as high risk (no information on e.g. registry, frequency of follow‐up) |
| Domain 4: Analysis | No | Dichotomisation of predictors. Although the choice of predictors was based on a previous model (CLL‐IPI), no formal factor selection procedure or comparison of tested factor combinations were reported. One point assigned per factor, no formally established factor weights |
| Overall judgement | No | |
Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants and number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding and conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | No recruitment period and study design reported No clear inclusion and exclusion criteria (we do not know if missing values have been part of the exclusion criteria) |
| Domain 2: Predictors | Yes | Not explicitly stated, but predictors probably assessed in a similar way |
| Domain 3: Outcome | Yes | No clear description of outcome assessment. We assumed that assessment of the objective outcome OS was similar within this single‐centre study and therefore did not rate as high risk (no information on e.g. registry, frequency of follow‐up) |
| Domain 4: Analysis | Unclear | No information on missing values; information on calibration and discrimination was provided upon request |
Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants and number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding and conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used. Many missing values due to retrospective design, rated in domain 4 |
| Domain 2: Predictors | Yes | Quote: "IgHV mutation analysis and FISH were performed at the reference laboratory of each participating center. The IgHV mutation status was tested on tumour DNA collected at diagnosis, and was assessed according to the ERIC guidelines." |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment. We assumed that assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | > 70% of individuals were excluded based on missing values (quote: "because of the absence of the required laboratory data in the cohort initially considered. However, the individuals included were representative of the whole cohort and were similar for age, sex, and Rai stage distribution"). |
| Overall judgement | No | |
Barcelona‐Brno V ‐ Molica 2017 (O‐CLL1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | Not applicable; outcome did not match primary outcome of the model | |
| Notes |
Funding & conflict of interest
Other comments
|
|
Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Although individuals with incomplete data were excluded, there was a comparison and the sample seemed representative of the cohort. |
| Domain 2: Predictors | Yes | The type of biomarkers used seem to be standard, and although there was no explicit information we think that the methods were consistent across studies and models. |
| Domain 3: Outcome | Yes | Objective standard outcome ‐ no clear description of outcome assessment ‐ we assumed that assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | Consecutive sampling of individuals in hospital routine, therefore around 2/3 of individuals with missing data for this model validation were excluded; no calibration reported. |
| Overall judgement | No | |
Barcelona‐Brno V ‐ Rani 2018 (Indian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Inclusion and exclusion criteria unclear; it seemed to us that only individuals with complete information on all predictors were included. Recruitment period unclear |
| Domain 2: Predictors | Yes | Not explicitly stated, but predictors probably assessed in a similar way (single‐centre) |
| Domain 3: Outcome | Unclear | Observation time to observe survival was short. |
| Domain 4: Analysis | No | Low number of events, no information on handling of missing values; calibration was not reported. |
| Overall judgement | No | |
Barcelona‐Brno V ‐ Reda 2017 (Milan cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | No eligibility criteria, unclear study design |
| Domain 2: Predictors | Yes | Not explicitly stated, but predictors probably assessed in a similar way |
| Domain 3: Outcome | Yes | No clear description of outcome assessment ‐ we assumed that assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | No information on number of events; no performance measures reported; patients with missing data were left out from analysis. |
| Overall judgement | No | |
CLL‐IPI D ‐ Bahlo 2016 (development cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used, missing values were excluded from analysis. This was rated in domain 4. |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way, according to RCT protocols. |
| Domain 3: Outcome | Yes | Objective standard outcome, follow‐up in the context of each individual RCT Median observation time seemed on the lower limit (around 5 years), but reasonable for patients with treatment indication. |
| Domain 4: Analysis | No | It was unclear for which model the performance was assessed (score or formula). Predictors were categorised and dichotomised. Patients with missing values were dropped from the analysis ‐ exclusion of approximately half of all patients. Univariable analysis was used for predictor selection. Upon request, the authors provided information on the calibration of their model. |
| Overall judgement | No | |
CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | No information on inclusion criteria, definition of CLL and if/how many patients were excluded due to missing baseline data |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way (single‐centre) |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment ‐ we assumed that assessment of the objective outcome OS was similar and therefore did not rate as high risk (no info on e.g. registry, frequency of follow‐up etc.) |
| Domain 4: Analysis | Yes | Number of events sufficient Patients with missing values possibly dropped at enrolment, not clearly stated: 'There were no missing values for the MAYO cohort.'. This was rated in domain 1. Upon request, the authors provided information on the calibration of their model. |
| Overall judgement | No | |
CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | The SCALE study database encompassed the entire population in Denmark and Sweden in a specified time frame. For this model validation, only the CLL cohort was relevant. We did not know exactly how many participants were excluded. No eligibility criteria were defined. |
| Domain 2: Predictors | Yes | Patient data collection between 1999 and 2002; we assumed that within this time frame, predictor assessment remained relatively homogenous. |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment in this publication. However, this was a national cohort study, therefore we assumed standardised outcome assessment. |
| Domain 4: Analysis | Yes | Sufficient number of events Missing data was handled appropriately: 'we analysed missing values of the SCAN cohort using Little’s MCAR test and imputed these using linear regression.' Upon request, the authors provided information on the calibration of their model. |
CLL‐IPI V ‐ Da Cunha‐Bang 2016 (Danish cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Individuals with missing data previously excluded; comparison of included and excluded individuals showed similar baseline characteristics. |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way. Relatively recent and prospectively followed cohort |
| Domain 3: Outcome | Unclear | For mortality, the observation time was too short (our clinician recommended observation time to exceed 5 years, median observation time here: 3.2 years) |
| Domain 4: Analysis | Unclear | Patients with missing data previously excluded, although authors stated that they were comparable. Rated in domain 1. No information on model performance measures, however, a validation in our sense was not planned. |
CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Retrospective retrieval of persons with available information for their model ‐ individuals without the necessary data may have been left out. Unclear recruitment period |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way (single‐centre study) |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment in this publication. We assumed standardised outcome assessment (single‐centre study). |
| Domain 4: Analysis | Yes | Sufficient number of events. No missing values, however, possibly an exclusion reason. This was rated in domain 1. Upon request, the authors provided information on the calibration of the CLL‐IPI in their cohort. |
| Overall judgement | No | |
CLL‐IPI V ‐ Gentile 2016 (Italian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used: consecutive sampling, but exclusion of individuals without baseline characteristics and FISH analysis from analysis (in total, only 22.5% of individuals were evaluable). This is rated in domain 4. |
| Domain 2: Predictors | Unclear | Due to the broad time range and multiple participating centres, we doubt that predictor assessments were homogeneous. |
| Domain 3: Outcome | Yes | Objective standard outcome: '... death, which were abstracted from clinical records at the time of inclusion and updated on an ongoing basis.' |
| Domain 4: Analysis | No | Consecutive sampling, but exclusion of individuals without baseline characteristics and FISH analysis from analysis (in total, only 22.5% of patients were evaluable). Missing values emerged due to the long time span. Some predictors were not assessed by default at diagnosis until recently. Comparison of baseline sample and included sample similar. Calibration not reported, however, survival per risk group available |
| Overall judgement | No | |
CLL‐IPI V ‐ Molica 2016 (O‐CLL1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | Not applicable; outcome did not match primary outcome of the model. | |
| Notes |
Funding & conflict of interest
Other comments
|
|
CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data source used: 'A total of 696 unselected CLL patients newly diagnosed and previously untreated from different institutions of the central region of Spain were included in this study.' |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment in this publication. We assume standardised outcome assessment. |
| Domain 4: Analysis | No | Insufficient number of events Participants with missing values not included in model ‐ consecutive sampling of routine patients, therefore around 2/3 of patients with missing data Calibration not reported |
| Overall judgement | No | |
CLL‐IPI V ‐ Rani 2018 (Indian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Inclusion and exclusion criteria unclear, it seemed like only patients with complete datasets were included. The recruitment period unclear |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way (single‐centre study). |
| Domain 3: Outcome | Unclear | Observation time was short. |
| Domain 4: Analysis | No | Low number of events; no information on handling of missing values; calibration was not reported. |
| Overall judgement | No | |
CLL‐IPI V ‐ Reda 2017 (Milano cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | No eligibility criteria reported |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way (single‐centre study) |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment in this publication. We assumed standardised outcome assessment |
| Domain 4: Analysis | No | Unclear how many events and how many patients etc. were included for validation Patients with missing values were excluded from analysis. No performance measures and survival per group reported (figure 1 not sharp) |
| Overall judgement | No | |
CLL‐IPI V ‐ Rigolin 2017 (Ferrera cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)rri
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | No eligibility criteria |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way (single‐centre study) |
| Domain 3: Outcome | Unclear | Objective standard outcome, however, observation time not reported |
| Domain 4: Analysis | No | No information on number of events, missing data and missing data handling; no performance measures reported |
| Overall judgement | No | |
CLL‐IPI V ‐ Zhu 2018 (Chinese cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | No eligibility criteria reported, unclear if patients with missing values were excluded |
| Domain 2: Predictors | Yes | Well‐established predictors used ‐ not explicitly stated, but predictors probably assessed in a similar way |
| Domain 3: Outcome | Yes | Objective standard outcome |
| Domain 4: Analysis | No | Number of events low (46) No information on missing values The authors provided the calibration and discrimination upon request. |
| Overall judgement | No | |
GCLLSG D ‐ Pflug 2014 (GCLLSG).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included persons in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used: combination of three RCT cohorts |
| Domain 2: Predictors | Yes | Not explicitly stated in this publication, but we assumed standard assessment in the context of each RCT |
| Domain 3: Outcome | Yes | Objective standard outcome, follow‐up in the context of each individual RCT |
| Domain 4: Analysis | No | Univariable selection of predictors Bootstrapping, however only for multivariable modeling and not used for correction of optimism; complete case analysis, background sample similar but many missing values; calibration not reported; simplification of the model, thus the assigned weights did not correspond to the results of multivariable modeling. |
| Overall judgement | No | |
GCLLSG V ‐ Molica 2015 (O‐CLL1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | Not applicable; outcome did not match primary outcome of the model. | |
| Notes |
Funding & conflict of interest
Other comments
|
|
GCLLSG V ‐ Pflug 2014 (Mayo cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Only patients with available data included: "... who had baseline data on all considered variables except s‐TK and/or s‐b2m available and who had stored serum collected > 36 months ..." |
| Domain 2: Predictors | Yes | Standard laboratory measures used. Where s‐TK and s‐B2M were not available, blood samples were shipped to Germany and analysed. |
| Domain 3: Outcome | Yes | Objective standard outcome ‐ no clear description of outcome assessment ‐ we assumed that assessment of the objective outcome OS was similar and therefore did not rate as high risk (no info on e.g. registry, frequency of follow‐up, etc.) |
| Domain 4: Analysis | No | Low number of events; patients with missing data were not enrolled in the study, rated in domain 1; calibration not reported |
| Overall judgement | No | |
GCLLSG V ‐ Rani 2018 (Indian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | Not applicable, outcome did not match primary outcome of the model | |
| Notes |
Funding & conflict of interest
Other comments
|
|
GIMEMA D ‐ Molica 2005 (GIMEMA cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used (retrospective cohort); 1.7% of patients were excluded due to inadequate follow‐up. We rated this as low proportion of exclusions. |
| Domain 2: Predictors | Yes | The predictors used seem to be routine demographic and laboratory information, and although there was no explicit information, we think that the methods were consistent. |
| Domain 3: Outcome | No | Knowledge of the predictor was used both in the definition of progression and the model (peripheral blood lymphocytes). |
| Domain 4: Analysis | No | Individuals with missing data were excluded: "The outcome of the 593 patients with complete data was different from the outcome of the 545 patients with incomplete data (10 year PFS 55.5% vs. 70.2%). The reasons for this difference rely on the higher number of patients in Rai stages I – III (P = 0.01) and increased serum levels of b2‐m (P = 0.003) found in the subgroup with complete data"; univariable selection of predictors; no weighting of predictors: "These differences in the RR did not prevent us from constructing a risk score simply by adding the negative factors present in a single patient at the time of diagnosis."; no model performance measures reported |
| Overall judgement | No | |
GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used: retrospective cohort |
| Domain 2: Predictors | Yes | The predictors used seem to be routine demographic and laboratory information, and although there was no explicit information, we think that the methods are consistent. |
| Domain 3: Outcome | Unclear | No median observation time; standard outcome, but treatment indication not described |
| Domain 4: Analysis | No | Only small amount of missing outcome data (n = 8); discrimination reported in form of AUC, however, time point not clear (probably 5 years); calibration not reported |
| Overall judgement | No | |
MDACC 2007 D ‐ Wierda 2007 (MDACC).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Retrospective cohort, consecutive sampling |
| Domain 2: Predictors | Yes | Routine clinical data used: although the recruitment period was very broad, standard clinical data (gender, age, Rai stage, ALC and beta‐2 microglobulin) have been included for modelling |
| Domain 3: Outcome | Yes | Objective standard outcome: no clear description of outcome assessment; we assumed that assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | Yes | Number of missing values and therefore the number of patients excluded from analysis was low; continuous factors handled appropriately; performance measures of original model reported; bootstrap correction applied; original formula and a simplified score provided; univariable selection of factors, however, nearly all factors were included in multivariable modelling as well. |
| Overall judgement | Yes | |
MDACC 2007 V ‐ Bulian 2011 (Italian‐Swiss).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | "Clinical and biological data of 1480 untreated CLL patients were retrospectively collected from 8 centres. The series included all cases observed at each centre over a given time period, ..." |
| Domain 2: Predictors | Yes | Measurements were taken in 8 different centres. However, the authors standardised the predictors so this should not be of influence. |
| Domain 3: Outcome | Yes | Objective standard outcome: no definition of outcome and assessment ‐ we assumed that definition and assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | Exclusion of individuals with missing values: "Some CLL cases with missing values were excluded from multivariate models. The death rate in this group was higher. The lack of inclusion of a number of individuals with possible worse prognosis could partly justify the observed bias in nomogram prediction."; expected OS estimated, but only point score: no discrimination calculated, because estimation based on nomogram not possible (quality of graph) |
| Overall judgement | No | |
MDACC 2007 V ‐ Gentile 2014 (Italian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Retrospective cohort: "... which included all patients diagnosed with CLL since 1983, were utilized for research purposes." |
| Domain 2: Predictors | Yes | Routine clinical data used: although the recruitment period was very broad, standard clinical data (gender, age, Rai stage, ALC and beta‐2 microglobulin) have been included for modelling. |
| Domain 3: Outcome | Yes | Objective standard outcome: no definition of outcome and assessment ‐ we assumed that definition and assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | Individuals with missing values were excluded. Although background characteristics were similar, we rated this as high risk because nearly half of all patients were excluded from analysis; measure for calibration not reported, however, reporting of survival per risk group |
| Overall judgement | No | |
MDACC 2007 V ‐ Gentile 2016 (Mayo cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Missing values as exclusion reason: 'The validation data set consisted of a consecutive series of 506 newly‐diagnosed CLL patients, prospectively followed at the Mayo Clinic, with baseline data available for all variables considered for both CLL‐IPI and the MDACC score.' |
| Domain 2: Predictors | Yes | Routine clinical data used, prospective follow‐up, relatively recent cohort (2001‐2008) |
| Domain 3: Outcome | Yes | Objective standard outcome, clear definition and assessment: 'death, which were abstracted from clinical records at the time of inclusion and updated on an ongoing basis.' |
| Domain 4: Analysis | Unclear | Individuals with missing data were excluded from the study, rated in domain 1. Calibration not reported; survival per risk groups was available. |
| Overall judgement | No | |
MDACC 2007 V ‐ González Rodríguez (Cabueñes).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Retrospective cohort, consecutive sampling: 'Un total de 265 pacientes se diagnosticaron de LLCB en el periodo de 10 anos.' |
| Domain 2: Predictors | Yes | Routine clinical data assessed at diagnosis |
| Domain 3: Outcome | Yes | Objective standard outcome: no definition of outcome and assessment ‐ we assumed that definition and assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | Low number of events; individuals with missing values excluded from modelling, however this number was small; calibration not reported |
| Overall judgement | No | |
MDACC 2007 V ‐ Molica 2010 (GIMEMA cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Retrospective cohort, inclusion of complete database: "The GIMEMA CLL database includes information on previously untreated CLL patients in Binet stage A whose diagnosis was immunologically confirmed". |
| Domain 2: Predictors | Yes | Although individuals came from 25 different centres, predictors were routine clinical data. |
| Domain 3: Outcome | Unclear | The median observation time was below 5 years (41.5 months); outcome time‐to‐first‐treatment was not the primary outcome of the validated model. |
| Domain 4: Analysis | No | Individuals with missing values were excluded from the analysis ("The outcome of the 593 patients with complete data was different from the outcome of the 545 patients with incomplete data"); no performance measures reported |
| Overall judgement | No | |
MDACC 2007 V ‐ Molica 2015 (O‐CLL1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | Not applicable, outcome did not match primary outcome of the model. | |
| Notes |
Funding & conflict of interest
Other comments
|
|
MDACC 2007 V ‐ Muñoz‐Novas 2018 (Spanish cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Retrospective cohort: "A total of 696 unselected CLL patients newly diagnosed and previously untreated from different institutions of the central region of Spain were included in this study." Patients with missing values were later excluded at analysis stage, rated in domain 4. |
| Domain 2: Predictors | Yes | Routine clinical data assessed at diagnosis was used (from medical records). |
| Domain 3: Outcome | Yes | Objective standard outcome: no definition of outcome and assessment ‐ we assumed that definition and assessment of the objective outcome OS was similar and therefore did not rate as high risk. |
| Domain 4: Analysis | No | Low number of event; exclusion of patients with missing values from analysis; calibration not reported |
| Overall judgement | No | |
MDACC 2007 V ‐ Pflug 2014 (3 RCTs).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Overlap of individuals between trials prevented; only concern could be comparability of the three trials. |
| Domain 2: Predictors | Yes | Prospective collection of predictors within clinical trials |
| Domain 3: Outcome | Yes | Objective standard outcome: follow‐up in frame of each RCT |
| Domain 4: Analysis | No | Number of events sufficient; individuals with missing data were excluded from model development. It was not clear if this was the case for the validation of the MDACC2007 model; calibration not reported |
| Overall judgement | No | |
MDACC 2007 V ‐ Rani 2018 (Indian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | No clear eligibility criteria; possibly missing values as exclusion reason; no recruitment period |
| Domain 2: Predictors | Yes | Study design unclear, "... were obtained from medical records of the patients". Routine clinical predictors were used in this model, therefore we rated this study as low. |
| Domain 3: Outcome | Unclear | Short follow‐up time |
| Domain 4: Analysis | No | Low number of events; calibration not reported; survival per risk groups graphically presented |
| Overall judgement | No | |
MDACC 2007 V ‐ Trajkova 2013 (Macedonia).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | "100 consecutive treatment naïve CLL patients" |
| Domain 2: Predictors | Yes | Routine clinical data |
| Domain 3: Outcome | Unclear | No observation time reported; outcome assessment not described |
| Domain 4: Analysis | No | Low number of participants and events; not really validating MDACC 2007 ‐ reporting of projected survival only and additional univariable analysis |
| Overall judgement | No | |
MDACC 2011 D ‐ Wierda 2011 (MDACC).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Individuals without evaluation of clinical factors and at least one newer factor were excluded: "We identified 930 previously untreated patients who presented to MD Anderson Cancer Center (MDACC) between January 2004 and December 2009, were not recommended for first‐line treatment at initial visit, and were evaluated for traditional clinical and laboratory prognostic factors and one or more of the newer prognostic factors including IgHV mutation status, chromosome abnormalities by FISH analysis, and ZAP‐70 expression by flow cytometry and/or immunohistochemistry (IHC)." |
| Domain 2: Predictors | Yes | Predictor assessment well described; comparison of assessment tools |
| Domain 3: Outcome | No | Some univariably tested predictors were not excluded from the outcome definition (progressive or symptomatic splenomegaly, massive nodes, progressive or symptomatic lymphadenopathy, Rai stage; Hallek 2008); short observation time |
| Domain 4: Analysis | No | Many predictors compared to events, 193 events (more than 25 predictors); univariable selection of predictors; unclear how many predictors entered into MV analysis; no performance measures reported; split sample, but after the model was already developed, which means that the data were internally validated on data that it was developed with. Bootstrapping was done, however not used to correct the model; assumption of proportionality was checked |
| Overall judgement | No | |
MDACC 2011 V ‐ Molica 2016 (O‐CLL1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Prospective cohort, detailed eligibility criteria in trial registry |
| Domain 2: Predictors | Yes | Predictors prospectively collected |
| Domain 3: Outcome | No | See development of the model: some predictors in the model were not excluded from the outcome definition (number and size of lymph nodes included in model, however these factors also formed Binet stage, which was a reason for treatment indication, Hallek 2008). |
| Domain 4: Analysis | No | Low number of events; no information on missing data reported |
| Overall judgement | No | |
Morabito D ‐ Morabito 2009 (Italian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Only one statement on inclusion criteria. No statement on baseline sample and possible exclusions. Very little baseline information on participants |
| Domain 2: Predictors | Yes | Prospective collection of predictor information, centralised laboratory |
| Domain 3: Outcome | Yes | Standard outcome definition used: "Treatment was decided uniformly in all participating centres based on documented progressive and symptomatic disease according to NCI working guidelines". |
| Domain 4: Analysis | No | Not sufficient events; continuous data not handled appropriately (ROC to find the 'best' cut‐point); no information on missing data; no performance measure reported |
| Overall judgement | No | |
Morabito V ‐ Gentile 2014 (O‐CLL1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Prospectively registered study (O‐CLL1‐GISL) with defined inclusion and exclusion criteria |
| Domain 2: Predictors | Yes | Prospective collection of predictor information. Predictors assessed study entry at a maximum of 1 year after diagnosis. |
| Domain 3: Outcome | Yes | CLL progression criteria 2008 were used. Outcome labelled differently than model development study. However, progression‐free survival (PFS) has been defined as time‐to‐therapy requirement, which is equivalent to time‐to‐treatment in Morabito 2009. |
| Domain 4: Analysis | Unclear | No relevant performance measures reported (progression per risk group was reported) |
O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Prospective cohort, clear inclusion and exclusion criteria in trial registry |
| Domain 2: Predictors | Yes | Prospective and standardised collection of predictor information ("CLL cell phenotype, CD38 and ZAP‐70 expression, and IgHV mutational status were performed in a central laboratory in Genova, while all FISH and genetic analyses were performed in Milan"). |
| Domain 3: Outcome | No | Standard outcome definition, however, predictors were not excluded from outcome definition (e.g. Rai stage). |
| Domain 4: Analysis | No | Continuous data not handled appropriately ("Continuous variables of prognostic importance on TFS in univariate proportional hazards Cox regression were dichotomized using published thresholds and laboratory norms"). |
| Overall judgement | No | |
O‐CLL‐1 V ‐ Rani 2018 (Indian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Inclusion and exclusion criteria unclear; it seemed to us that only individuals with complete information on all predictors were included; recruitment period unclear; study design unclear |
| Domain 2: Predictors | Yes | Not explicitly stated, but predictors probably assessed in a similar way (single‐centre) |
| Domain 3: Outcome | No | See model development: Predictors were not excluded from outcome definition (e.g. Rai stage). |
| Domain 4: Analysis | No | Low number of events; no information on handling of missing values |
| Overall judgement | No | |
O‐CLL1 V ‐ Gentile 2016 (Mayo cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | No eligibility criteria; individuals with missing values were excluded. |
| Domain 2: Predictors | Yes | Not explicitly stated, but predictors probably assessed in a similar way (single‐centre). |
| Domain 3: Outcome | No | See model development: Predictors not excluded from outcome definition (e.g. Rai stage) |
| Domain 4: Analysis | Unclear | Calibration not reported; individuals with missing values were probably excluded from analysis, rated in domain 1. |
| Overall judgement | No | |
Rossi D ‐ Rossi 2013 (Italian cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Yes | Appropriate data sources used: "multicentric cohort of 637 newly diagnosed and previously untreated CLL patients who consecutively presented for initial evaluation at four institutions from June 1996 through June 2011" |
| Domain 2: Predictors | Yes | Detailed description of predictors and timing of measurements: "clinical information prospectively collected at clinically relevant time points (i.e. at diagnosis, progression, and last follow‐up)" |
| Domain 3: Outcome | Yes | Objective standard outcome; regular clinical database update |
| Domain 4: Analysis | Unclear | Unclear if individuals with missing data were excluded; number of events sufficient; building of model by recursive partitioning (decision tree, tenfold cross‐validation), testing for stability with random forest algorithm and Cox model with bootstrapping; correction of c‐statistic; assumptions were checked |
Rossi V ‐ Jeromin 2014 (Munich cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Study design unclear |
| Domain 2: Predictors | Yes | Relatively short recruitment period (2005 ‐ 2010); samples were sent to a laboratory which was not involved in treatment. |
| Domain 3: Outcome | Yes | Objective standard outcome; no information on outcome assessment. However, we assumed that for OS, risk of bias was low. |
| Domain 4: Analysis | No | Individuals with missing data excluded from analysis (complete data for OS in 935/1160 cases); no performance measures, just risk groups |
| Overall judgement | No | |
Rossi V ‐ Rossi 2013 (unclear).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | Unclear | Study design not clear: "The validation series was represented by a cohort of 370 newly diagnosed and previously untreated CLL patients who consecutively presented for initial evaluation from June 1996 through June 2011 (Table S1) and provided with regular follow‐up (at least three visits/year)." |
| Domain 2: Predictors | Yes | No detailed description of predictor assessment; we assumed that it was similar to the development cohort described in the same publication. |
| Domain 3: Outcome | Yes | Objective standard outcome, regular clinical database update |
| Domain 4: Analysis | No | Small number of events; no information on missing values; calibration not reported |
| Overall judgement | No | |
Stephens OS D ‐ Stephens 2015 (Ohio cohort).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
Number of candidate predictors
List of predictors in final model (including cut‐points for dichotomised factors)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Predictor selection method
Statistical method
Simplification of model?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Possibly selective inclusion of individuals, as only patients with del(17p13.1) were considered. Individuals without assessment could not have been included. Other eligibility criteria were unclear. |
| Domain 2: Predictors | Yes | Mostly laboratory data, probably assessed in a similar way: "Stimulated cytogenetic and fluorescent in situ hybridisation (FISH) analyses were performed on peripheral blood or bone marrow samples, as previously described"; single‐centre study |
| Domain 3: Outcome | Yes | Objective standard outcome: although median observation time was not reported, median OS estimates were 5.2 years, therefore, we assumed a median observation time exceeding 5 years. |
| Domain 4: Analysis | No | Number of events only reported for TFS, not OS ‐ the number of persons included was too small, total n = 114; missing data handling appropriate; inconsistent reporting of c‐statistics; calibration was not reported |
| Overall judgement | No | |
Stephens OS V ‐ Stephens 2015 (MDACC).
| Study characteristics | ||
| General information |
Model and type of study
Secondary citations
Language of publication
Study design
Follow‐up time
|
|
| Participants |
Number of included participants in the cohort
Setting
Recruitment period
Age (in years)
Sex
Stages of disease
Treatment
Inclusion criteria
Exclusion criteria
|
|
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
Timing of predictor measurement
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data |
Participants with any missing data?
If yes, how was missing data handled?
|
|
| Analysis |
Number of participants & number of events (specific time points where reported)
Which model was used?
Was the model updated?
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability |
Domain 1: Participant selection
Domain 2: Predictors
Domain 3: Outcome
|
|
| Notes |
Funding & conflict of interest
Other comments
|
|
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Possibly selective inclusion of individuals, as only patients with del(17p13.1) were considered. Individuals without assessment could not have been included. Other eligibility criteria were unclear. |
| Domain 2: Predictors | Unclear | lack of information about similarity of predictor assessment to the development study; no recruitment period |
| Domain 3: Outcome | Yes | Objective standard outcome: although median observation time was not reported, median OS estimates were 6.4 years, therefore we assumed a median observation time exceeding 5 years. |
| Domain 4: Analysis | No | Number of included persons too small, total N = 129; calibration not reported |
| Overall judgement | No | |
Stephens TFS D ‐ Stephens 2015 (Ohio cohort).
| Study characteristics | ||
| General information |
Model and type of study
Different outcome but identical publication as Stephens OS D ‐ Stephens 2015 (Ohio cohort) and Stephens OS V ‐ Stephens 2015 (MDACC). Therefore, only relevant differences are reported in this table. For additional information, see tables above. |
|
| Participants | see Stephens OS D ‐ Stephens 2015 (Ohio cohort) | |
| Predictors |
List of predictors in final model (including cut‐points for dichotomised factors)
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
|
|
| Missing data | ||
| Analysis |
Number of participants & number of events (specific time points where reported)
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | ||
| Notes | ||
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Possibly selective inclusion of individuals, as only patients with del(17p13.1) were considered. Individuals without assessment could not have been included. Other eligibility criteria were unclear. |
| Domain 2: Predictors | Yes | Mostly laboratory data, probably assessed in a similar way: "Stimulated cytogenetic and fluorescent in situ hybridization (FISH) analyses were performed on peripheral blood or bone marrow samples, as previously described"; single‐centre study |
| Domain 3: Outcome | No | Predictors were not excluded from the outcome definition (Rai stage; Hallek 2008) |
| Domain 4: Analysis | No | Number of events only reported for TFS, not OS ‐ the number of persons included was too small, total N = 114; missing data handling appropriate; inconsistent reporting of c‐statistics; calibration was not reported |
| Overall judgement | No | |
Stephens TFS V ‐ Stephens 2015 (MDACC).
| Study characteristics | ||
| General information |
Model and type of study
Different outcome but identical publication as Stephens OS D ‐ Stephens 2015 (Ohio cohort) and Stephens OS V ‐ Stephens 2015 (MDACC). Therefore, only relevant differences are reported in this table. For additional information, see tables above. |
|
| Participants | ||
| Predictors |
List of predictors used for validation (and changes between original predictors and predictors in validation study)
|
|
| Outcome(s) |
Primary outcome in study
Additional outcome(s)
Outcome in model development
|
|
| Missing data | ||
| Analysis |
Number of participants & number of events (specific time points where reported)
Performance measures reported?
Creation of risk groups?
|
|
| PROBAST: Applicability | ||
| Notes | ||
| Item | Authors' judgement | Support for judgement |
| Domain 1: Participant selection | No | Possibly selective inclusion of individuals, as only patients with del(17p13.1) were considered. Individuals without assessment could not have been included. Other eligibility criteria were unclear. |
| Domain 2: Predictors | Unclear | Lack of information about similarity of predictor assessment to the development study, no recruitment period |
| Domain 3: Outcome | No | Predictors were not excluded from the outcome definition (Rai stage; Hallek 2008) |
| Domain 4: Analysis | No | Number of included persons too small, total N = 129; calibration not reported |
| Overall judgement | No | |
AIHA: autoimmune haemolytic anaemia ALC: absolute lymphocyte count AUC: area under the curve B2M: beta2‐microglobulin BcR IG: clonotypic B‐cell receptor immunoglobulin BIRC3: baculoviral IAP repeat containing 3 BR: bendamustine‐rituximab C: cyclophosphamide CHL: chlorambucil CHOP: cyclophosphamide, doxorubicin, vincristine, and prednisone CI: confidence interval CIRS: Cumulative Illness Rating Score CLL: chronic lymphocytic leukaemia CLL‐IPI: Chronic Lymphocytic Leukaemia‐International Prognostic Index cMBL: clinical monoclonal B lymphocytosis CVP: central venous pressure DE: Germany del11q: FISH‐detected genetic abberation del11q del17p: FISH‐detected genetic abberation del17p ECOG PS: Eastern Cooperative Oncology Group performance status F: fludarabine FC: fludarabine and cyclophosphamide FCR: fludarabine, cyclophosphamide and rituximab FFP: fresh frozen plasma FR: France FISH: fluorescence in situ hybridisation HIV: human immunodeficiency virus HBV: hepatitis B virus HCV: hepatitis C virus HDMP: high‐dose methylprednisolone IgHV: immunoglobulin heavy chain variable region genes IGLV: immunoglobulin lambda variable cluster IQR: Interquartile range IPD: individual patient data iwCLL: International workshop on Chronic Lymphocytic Leukemia LDH: lactate dehydrogenase LDT: lymphocyte doubling time LN: lymph node LYM: lymphocyte count MCAR: missing completely at random M‐CLL: mutated chronic lymphocytic leukaemia MDACC: MD Anderson Cancer Center MLL: Munich Leukemia Laboratory MoAbs: monoclonal antibodies MV analyses: multivariable analysis NA: not available NCI: National Cancer Institute NOTCH1: notch receptor 1 O‐Benda: ofatumumab bendamustine O:E: observed‐expected ratio OS: overall survival PFS: progression‐free survival PL: Poland PROBAST: Prediction model Risk Of Bias ASsessment Tool R: rituximab RCT: randomised controlled trial ROC: receiver operating characteristics curve RR: risk ratio RS: risk score RTK: tyrosine kinase inhibitors s‐B2M: serum beta‐2 microglobulin SCALE: Scandinavian population‐based study SCAN: Scandinavian population‐based case‐control study SF3B1: Splicing Factor 3b Subunit 1 SRS: simplified risk score s‐TK: serum thymidine kinase TFS: treatment‐free survival TP53: tumour protein P53 TP: treatment probability TTE: Time‐to‐Evenr TTFT: time‐to‐first‐treatment U‐CLL: unmutated chronic lymphocytic leukaemia WBC: white blood cell count WHO: World Health Organization
Characteristics of excluded studies [ordered by study ID]
| Study | Reason for exclusion |
|---|---|
| Apelgren 2006 | Study focus on staging system(s) |
| Baccarani 1982 | Study focus on staging system(s) |
| Baliakas 2015 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Berke 2019 | Prognostic factor study |
| Bettini 1986 | Outdated staging system, which is no longer used in today's clinical practice |
| Binet 1977 | Study focus on staging system(s) |
| Binet 1981 | Study focus on staging system(s) |
| Bo 2014 | Prognostic factor study |
| Bomben 2005 | Comment or addition referring to an excluded study |
| Bomben 2009 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Bou Samra 2014 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Brejcha 2010 | Prognostic factor study |
| Brugiatelli 2007 | Prognostic factor study |
| Bulian 2014 | Prognostic factor study |
| Byrd 2006 | Prognostic factor study |
| Cailliod 2005 | Prognostic factor study |
| Callea 1999 | Prognostic factor study |
| Catovsky 1989 | Prognostic factor study |
| Cesano 2013 | Prognostic factor study |
| Chang 2003 | Prognostic factor study |
| Chastang 1985 | Outdated staging system, which is no longer used in today's clinical practice |
| Chauzeix 2018 | Prognostic factor study |
| Chelazzi 1979 | Study focus on staging system(s) |
| Chen 1997 | Prognostic factor study |
| Chena 2008 | Prognostic factor study |
| Chevallier 2002 | Prognostic factor study |
| Chiaretti 2014 | Prognostic factor study |
| Christiansen 1994 | Prognostic factor study |
| Chuang 2012 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Ciccone 2012 | Prognostic factor study |
| Ciocoiu 1988 | Study focus on staging system(s) |
| Claus 2012 | Prognostic factor study |
| Claus 2014 | Prognostic factor study |
| Cmunt 2002 | Prognostic factor study |
| Cocco 2005 | Prognostic factor study |
| Corcoran 2005 | Prognostic factor study |
| Cordone 1998 | Prognostic factor study |
| Cortese 2014 | Prognostic factor study |
| Coscia 2012 | Prognostic factor study |
| Crespo 2003 | Prognostic factor study |
| Criel 1997 | Characterisation of CLL and prognostic factors |
| Cro 2009 | Prognostic factor study |
| Cro 2010 | Characterisation of CLL and prognostic factors |
| Cuneo 2004 | Characterisation of CLL and prognostic factors |
| Cuneo 2018 | Observational efficacy study |
| D'Arena 2001 | Prognostic factor study |
| D'Arena 2007 | Prognostic factor study |
| D'Arena 2012 | Characterisation of CLL and prognostic factors |
| Damle 1999 | Prognostic factor study |
| Dasgupta 2015 | Study focus on identification of the threshold for a prognostic factor |
| Davis 2016 | Study focus on identification of the threshold for a prognostic factor |
| De Faria 2000 | Study focus on staging system(s) |
| De Rossi 1989 | Study focus on staging system(s) |
| DeAndres‐Galiana 2016 | Study focus on identification of a prognostic factor |
| Degan 2004 | Prognostic factor study |
| Del Guidice 2011 | Prognostic factor study |
| Del Poeta 2010 | Prognostic factor study |
| Del Principe 2004 | Prognostic factor study |
| Del Principe 2006 | Prognostic factor study |
| Delgado 2009 | Prognostic factor study |
| Delgado 2014 | Prognostic factor study |
| Deslandes 2007 | Methodological study |
| Di Raimondo 2001 | Prognostic factor study |
| Dimier 2018 | Prediction model to evaluate the relationship between surrogacy outcome(s) and standard outcomes |
| Dong 2011 | Prognostic factor study |
| Dong 2014 | Prognostic factor study |
| Durak 2009 | Prognostic factor study |
| El‐Kinawy 2012 | Prognostic factor study |
| Fang 2019 | Study applies the CLL‐IPI, but without the aim of validating it |
| Ferrara 1981 | Study focus on staging system(s) |
| Ferreira 2014 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| French Cooperative Group on CLL 1988 | Outdated staging system, which is no longer used in today's clinical practice |
| Friedman 2009 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Gattei 2008 | Prognostic factor study |
| Gdynia 2018 | Prognostic factor study |
| Geisler 1997 | Characterisation of CLL and prognostic factors |
| Gentile 2016 | Prognostic factor study |
| Gentile 2018 | Score or model included predictors available only at treatment initiation |
| Giles 2003 | Patient population did not match the review question; study included previously treated CLL patients |
| Giudice 2018 | Prognostic factor study |
| Gogia 2014 | Prognostic factor study |
| Gonzalez 2013 | Characterisation of CLL and prognostic factors |
| Gonzalez‐Gascon 2015 | Characterisation of CLL and prognostic factors |
| Gonzalez‐Rodriguez 2010 | Characterisation of CLL and prognostic factors |
| Grabowski 2005 | Prognostic factor study |
| Grever 2006 | Abstract only |
| Hallek 1999 | Characterisation of CLL and prognostic factors |
| Han 1984 | Prognostic factor study |
| Herold 2011 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Hock 2010 | Prognostic factor study |
| Houldsworth 2014 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Hus 2006 | Prognostic factor study |
| Jaksic 1981 | Prognostic factor study |
| Jaksic 1992 | Study focus on staging system(s) |
| Jaksic 2014 | Comment or addition referring to an excluded study |
| Josefsson 2007 | Prognostic factor study |
| Juliusson 1986 | Prognostic factor study |
| Juliusson 1990 | Prognostic factor study |
| Kahraman 2014 | Prognostic factor study |
| Kardum‐Skelin 2008 | Prognostic factor study |
| Kardum‐Skelin 2009 | Patient population did not match the review question; study included previously treated CLL patients |
| Karmiris 1994 | Prognostic factor study |
| Kay 2018 | Study applied the CLL‐IPI, but without the aim of validating it |
| Keating 2000 | Patient population did not match the review question; study included previously treated CLL patients |
| Khalifa 2002 | Prognostic factor study |
| Kim 2004 | Prognostic factor study |
| Kimby 1988 | Prognostic factor study |
| Kleinstern 2018 | Risk factor study |
| Knospe 1977 | Prognostic factor study |
| Koberda 1989 | Prognostic factor study |
| Korycka‐Wolowiec 2011 | Prognostic factor study |
| Krober 2002 | Prognostic factor study |
| Krober 2006 | Patient population did not match the review question; study included previously treated CLL patients |
| Kryachok 2011 | Prognostic factor study |
| Kurec 1992 | Prognostic factor study |
| Lai 2002 | Prognostic factor study |
| Lech‐Maranda 2012 | Prognostic factor study |
| Lech‐Maranda 2013 | Prognostic factor study |
| Lecouvet 1997 | Prognostic factor study |
| Li 2008 | Prognostic factor study |
| Li 2017a | Prognostic factor study |
| Lin 2002 | Prognostic factor study |
| Lin 2014 | Prognostic factor study |
| Lozano‐Santos 2014 | Prognostic factor study |
| Lucas 2015 | Prognostic factor study |
| Maffei 2007 | Prognostic factor study |
| Maffei 2010 | Prognostic factor study |
| Mandelli 1987 | Development study of an outdated staging system, which is no longer used in today's clinical practice |
| Mansouri 2013 | Prognostic factor study |
| Marasca 2005 | Prognostic factor study |
| Marasca 2013 | Prognostic factor study |
| Martinelli 2008 | Prognostic factor study |
| Masic 1998 | Prognostic factor identification study |
| Mateva 2001 | Prognostic factor model, no full‐text for further investigation |
| Matthews 2006 | Prognostic factor study |
| Matthews 2007 | Prognostic factor study |
| Matutes 2013 | Prognostic factor study |
| Matutes 2017 | Review |
| Melo 1987 | Patient population did not match the review question; study was not restricted to previously untreated CLL patients |
| Miao 2018 | Prognostic factor study |
| Molica 1984 | Study focus on staging system(s) |
| Molica 1986 | Prognostic factor study |
| Molica 1988 | Prognostic factor study |
| Molica 1991 | Prognostic factor study |
| Molica 1994 | Prognostic factor study |
| Molica 1998 | Prognostic factor study |
| Molica 1999a | Prognostic factor study |
| Molica 1999b | Prognostic factor study |
| Molica 2008 | Prognostic factor study |
| Montserrat 1991 | Prognostic factor study |
| Morabito 2001 | Prognostic factor study |
| Morabito 2015a | Prognostic factor study |
| Morabito 2015b | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Morabito 2018c | Study aimed to identify cut‐off points for a specific prognostic factor |
| Moreno 2019 | Study focus on staging system(s) |
| Morilla 2008 | Prognostic factor study |
| Nabhan 2017 | Score or model included predictors available only at treatment initiation |
| Nedeva 2018 | Evaluation of an excluded model |
| Nenova 2000 | Prognostic factor study |
| Nipp 2014 | Prognostic factor study |
| Nola 2004 | Patient population did not match the review question; study was not restricted to previously untreated CLL patients |
| Nowakowski 2009 | Prognostic factor study |
| Nuckel 2006 | Prognostic factor study |
| Nuckel 2009 | Prognostic factor study |
| O'Brien 1993 | Patient population did not match the review question; study was not restricted to previously untreated CLL patients |
| Ocana 2007 | Prognostic factor study |
| Oliveira 2011 | Prognostic factor study |
| Orgueira 2019 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Oscier 1990 | Prognostic factor study |
| Paolino 1984 | Prognostic factor study |
| Plesingerova 2017 | Study focus on surrogacy outcome(s) |
| Prokocimer 1985 | Prognostic factor study |
| Qin 2017 | Prognostic factor study |
| Queiros 2015 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Rai 1975 | Study focus on staging system(s) |
| Rai 1990 | Study focus on staging system(s) |
| Raponi 2018 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Resegotti 1989 | Prognostic factor study |
| Rissiek 2014 | Prognostic factor study |
| Rodriguez 2007 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Ronchetti 2016 | Prognostic factor study |
| Rossi 1986 | Study focus on staging system(s) |
| Rossi 2010a | Study focus on identification of the threshold for a prognostic factor |
| Rossi 2010b | Patient population did not match the review question; study was not restricted to previously untreated CLL patients |
| Rossi 2011 | Patient population did not match the review question; study was not restricted to previously untreated CLL patients |
| Rozman 1979 | Study focus on staging system(s) |
| Salomon‐Nguyen 1995 | Diagnostic score |
| Santoro 1979 | Study focus on staging system(s) |
| Sarmiento 2002 | Prognostic factor study |
| Savvopoulos 2016 | Methodological study |
| Scolozzi 1981 | Study focus on staging system(s) |
| Shanafelt 2010 | Prognostic factor study |
| Shanafelt 2017 | Outcome not relevant for the review |
| Spacek 2009 | Prognostic factor study |
| Stamatopoulos 2009 | Prognostic score, however, this score was built with a proxy as outcome ('distinguished mutated and unmutated cases') |
| Stamatopoulos 2017 | Prognostic factor study |
| Strefford 2015 | Prognostic factor study |
| Szymczyk 2018 | Prognostic factor study |
| Tallarico 2018 | Outcome not relevant for the review |
| Tobin 2005a | Study focus on identification of the threshold for a prognostic factor |
| Tobin 2005b | Study focus on identification of the threshold for a prognostic factor |
| Vallat 2013 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Van Damme 2012 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
| Velardi 1980 | Study focus on staging system(s) |
| Vojdeman 2017 | Prognostic factor study |
| Vural 2014 | Prognostic factor study |
| Weinberg 2007 | Patient population did not match the review question; study was not restricted to previously untreated CLL patients |
| Weiss 2011 | Prognostic factor study |
| Wierda 2003 | Prognostic factor study |
| Winkler 2010 | Prognostic factor study |
| Wu 2010 | Study focus on staging system(s) |
| Zengin 1997 | Study focus on staging system(s) |
| Zenz 2009 | Prognostic factor study |
| Zucchetto 2006 | Genetic analysis, e.g. genetic subgrouping, genetic signature(s) or genetic clustering |
CLL: chronic lymphocytic leukaemia CLL‐IPI: chronic lymphocytic leukaemia International Prognostic Index
Characteristics of studies awaiting classification [ordered by study ID]
Antic 2011.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Baumann 2014.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Bulian 2011.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Bulian 2012.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Cavallini 2017.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Del Guidice 2005.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Friedrichs 2011.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Furundarena 1994.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Gentile 2009.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Haferlach 2010.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Jarque 1991.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Lee 1987.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Leotard 2000.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Letestu 2010.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Li 2017b.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Liang 2018.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Metze 2000.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Miao 2019.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Molica 1990.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Molica 2010.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Molica 2015.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Molica 2019.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Morabito 2011.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Pepper 2012.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Qin 2018.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Rozman 1982.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Rozman 1984.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Schweighofer 2011.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Stamatopoulos 2010.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Tsimberidou 2007.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Vetro 2018.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Visentin 2015.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Wierda 2009.
| Notes | Details on study design, recruitment period, prognostic factors and more can be found in Appendix 7. |
Characteristics of ongoing studies [ordered by study ID]
NCT00275054.
| Study name | Rituximab, fludarabine, and cyclophosphamide or observation alone in treating patients with stage 0, stage I, or stage II chronic lymphocytic leukemia |
| Starting date | 2005 |
| Contact information | Michael Hallek, MD, Medizinische Universitaetsklinik I at the University of Cologne |
| Notes |
|
NCT03436524.
| Study name | A prognostic tool for early stage CLL |
| Starting date | 2018 |
| Contact information | Davide Rossi, MD, PhD, Principal investigator, Oncology Institute of Southern Switzerland |
| Notes |
|
Differences between protocol and review
We changed the title of the review to clarify the included population (newly‐diagnosed adults with CLL). In the protocol, the title was 'Prognostic models for chronic lymphocytic leukaemia: an exemplar systematic review and meta‐analysis'.
Structure of the review
As prognosis reviews are a new review type within Cochrane, little guidance had been published at protocol stage. Recently, a review template has been developed and published by the Cochrane Prognosis Methods group, which we have adopted for this review (methods.cochrane.org/prognosis/our-publications). In addition, we have added two sections ('Selection of studies' and ''Risk of bias' assessment') to report our group decisions transparently.
Search methods
We did not search PubMed, because content from PubMed can be identified via MEDLINE.
We added a search for Embase based on editorial comments.
We added a search of the following databases for ongoing trials instead of the metaRegister of Controlled Trials:
ClinicalTrials.gov
World Health Organization International Clinical Trials Registry Platform (WHO ICTRP)
We did not search conference proceedings for abstracts, because the paucity of information would not allow us to apply the 'Risk of bias' tool. To ensure that we identified all validation studies corresponding to a developed model, we screened the citations of all included studies (Web of Science, October 2018).
Inclusion and exclusion of studies
Originally, we planned to include only models published after 1990. However, prognostic model studies are often conducted based on retrospective data, which sometimes include the analysis of blood samples frozen at diagnosis and analysed many years after. To give the same chance to studies with samples that were analysed immediately and after several years for inclusion in this review, we decided not to limit our search strategy based on date of publication. As a clarification, any studies that explicitly aimed at defining a new staging system was excluded during title and abstract screening.
We decided to include prognostic models with the outcomes 'time‐to‐first treatment' and 'treatment‐free survival' as additional outcomes, because individuals with low‐risk CLL can often live a long time without disease progression and the need for treatment. Usually, at disease progression, treatment is indicated, which means that these two outcomes are quite similar and both are meaningful for the patient.
After several rounds of discussion, we decided that genetic signatures are beyond the scope of this review because: a) we prespecified that we would exclude factor identification studies, which most of the genetic signature studies are to start with; b) they are not yet ready to be applied in clinical practice due to the unavailability of detailed genetic information that these signatures use and the complexity of the algorithms; and c) more often than not, it is not feasible to assess a genetic signature study in the same context as a prognostic model.
Analysis and 'Risk of bias' assessment
Instead of the CHARMS checklist, we used the recently published PROBAST‐tool to assess the risk of bias of individual studies (Wolff 2019). The checklist does not originally aim at the assessment of risk of bias, but as a guide for critical appraisal and data extraction. However, at the protocol stage, no tool specific to 'Risk of bias' assessment for prognostic models had been published.
The analysis was not specified precisely in the protocol, as methods for systematic reviews of prognosis are just developing. As described in the protocol, we decided to follow the current recommendations of the Cochrane Prognosis Methods Group. We added a more detailed description in the section Data collection and analysis. In short, we meta‐analysed the performance measures of the various external validation studies per model where data were available. We planned to summarise the measures of calibration. Unfortunately, calibration in the form of O:E ratios or calibration plots was rarely reported. Instead, many studies only presented the observed outcome frequency per subgroup at a specific time point. To gain an overview of this information among several external validations, during the review process and in collaboration with the Cochrane Prognsosis Methods Group, we decided to represent the outcome frequency graphically together with the pooled outcome frequency in a table.
As we only meta‐analysed the external validation studies of a prognostic model, we analysed only models that were externally validated several times. Although we have included all prognostic models and scores that we identified, we did not describe models without any external validation studies in detail, because it is not recommended to use prognostic scores or models without any testing in independent cohorts, especially when the development sample was small (Moons 2015; Steyerberg 2013). References to these studies can be found in the list of studies awaiting classification; further information can be found in Appendix 7.
In the protocol, we did not specify any sensitivity analyses. We decided post hoc to explore the effect of including area under the curve (AUC) as a performance measure for discrimination instead of the c‐statistic (Figure 23), the effect of the Newcombe estimation method for 95% CIs of the c‐statistic (Figure 24; Figure 25), and the difference between studies using the original predictor or a proxy (del(17p) instead of TP53 mutation (Figure 26).
GRADE
We did not apply GRADE as no GRADE guidance has yet been developed to assess certainty of evidence from meta‐analysis of prognostic models.
Contributions of authors
Nina Kreuzberger: screening and selection of studies, development of data extraction form and data extraction, characteristics of studies, 'Risk of bias' assessment, statistical analysis, writing and drafting of the review, communication with and between authors
Johanna AAG Damen: statistical analysis
Marialena Trivella: statistical input, screening and selection of studies, data extraction, 'Risk of bias' assessment
Lise J Estcourt: medical and content input, screening and selection of studies, data extraction, 'Risk of bias' assessment
Angela Aldin: screening and selection of studies, data extraction, 'Risk of bias' assessment
Lisa Umlauff: data extraction, 'Risk of bias' assessment, characteristics of included studies
Maria Vazquez Montes: prototype of data extraction form, support in data extraction, 'Risk of bias' assessment
Robert Wolff: 'Risk of bias' input
Karel GM Moons: methodological input on reviews of prognosis studies
Ina Monsef: search strategy development
Farid Foroutan: 'Risk of bias' input
Karl‐Anton Kreuzer: medical and content input
Nicole Skoetz: protocol development, screening and selection of studies, data extraction, 'Risk of bias' assessment, extensive proofread and comments on the review draft
Sources of support
Internal sources
University Hospital of Cologne, Department I of Internal Medicine, Germany
NHS Blood and Transplant, UK
External sources
Grant by the Federal Ministry of Education and Research (Grant no. 01KG1711), Germany
Declarations of interest
Nina Kreuzberger: My institution received a grant from the Federal Ministry of Education and Research, Germany to conduct this review.
Johanna AAG Damen: none known
Marialena Trivella: I am working as a statistical editor in a number Cochrane groups, and I declare that my work as a statistical editor is independent to this published work where I participate. The University of Oxford received a small part of the grant from the Federal Ministry of Education and Research, Germany, as reimbursement for my time spent on the project.
Lise J Estcourt: My institution received a grant from the Federal Ministry of Education and Research, Germany to conduct this review.
Angela Aldin: My institution received a grant from the Federal Ministry of Education and Research, Germany to conduct this review.
Lisa Umlauff: My institution received a grant from the Federal Ministry of Education and Research, Germany to conduct this review.
Maria Vazquez Montes: I am sponsored by the BHF to contribute on a similar prognostic models review for heart failure which allowed me to effectively participate in the work under consideration.
Robert Wolff: As employee of Kleijnen Systematic Reviews, I was the lead author of a systematic review on prostate cancer, commissioned by Elekta, Nucletron.
Karel Moons: none known
Ina Monsef: none known
Farid Foroutan: none known
Karl‐Anton Kreuzer: board member and consultant for AbbVie, Alexion, Amgen, Ariad, Baxter, Bayer Health Care, Biotest, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Chugai, Gilead, Glaxo‐SmithKline, Grifols, Hexal, Janssen, Jazz Pharmaceuticals, Leo, Mundipharma, MSD, Novartis, Pfizer, Roche, Shire, Teva. Grants, fees, honoraria and travel grants from AbbVie, Alexion, Amgen, Ariad, Baxter, Bayer Health Care, Biotest, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Chugai, Gilead, Glaxo‐SmithKline, Grifols, Hexal, Janssen, Jazz Pharmaceuticals, Leo, Mundipharma, MSD, Novartis, Pfizer, Roche, Shire, Teva.
Nicole Skoetz: My institution received a grant from the Federal Ministry of Education and Research, Germany to conduct this review.
New
References
References to studies included in this review
Baliakas D ‐ Baliakas 2019 (multicentre) {published data only}
- Baliakas P, Moysiadis T, Hadzidimitriou A, Xochelli A, Jeromin S, Agathangelidis A, et al. Tailored approaches grounded on immunogenetic features for refined prognostication in chronic lymphocytic leukemia. Haematologica 2019;104(2):360-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Baliakas V ‐ Baliakas 2019 (MLL + Scan.) {published data only}
- Baliakas P, Moysiadis T, Hadzidimitriou A, Xochelli A, Jeromin S, Agathangelidis A, et al. Tailored approaches grounded on immunogenetic features for refined prognostication in chronic lymphocytic leukemia. Haematologica 2019;104(2):360-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Barcelona‐Brno D ‐ Delgado 2017 (Barcelona cohort) {published data only}
- Delgado J, Doubek M, Baumann T, Kotaskova J, Molica S, Mozas P, et al. Chronic lymphocytic leukemia: a prognostic model comprising only two biomarkers (IGHV mutational status and FISH cytogenetics) separates patients with different outcome and simplifies the CLL-IPI. American Journal of Hematology 2017;92(4):375-80. [DOI] [PubMed]
Barcelona‐Brno V ‐ Delgado 2017 (Brno cohort) {published data only}
- Delgado J, Doubek M, Baumann T, Kotaskova J, Molica S, Mozas P, et al. Chronic lymphocytic leukemia: a prognostic model comprising only two biomarkers (IGHV mutational status and FISH cytogenetics) separates patients with different outcome and simplifies the CLL-IPI. American Journal of Hematology 2017;92(4):375-80. [DOI] [PubMed]
Barcelona‐Brno V ‐ Gentile 2017 (Italian & Mayo) {published data only}
Barcelona‐Brno V ‐ Molica 2017 (O‐CLL1‐GISL) {published data only}
- Delgado J, Doubek M, Baumann T, Kotaskova J, Molica S, Mozas P, et al. Chronic lymphocytic leukemia: a prognostic model comprising only two biomarkers (IGHV mutational status and FISH cytogenetics) separates patients with different outcome and simplifies the CLL-IPI. American Journal of Hematology 2017;92(4):375-80. [DOI] [PubMed]
- *.Molica S, Giannarelli D, Mirabelli R, Levato L, Gentile M, Morabito F, et al. Reliability of six prognostic models to predict time-to-first-treatment in patients with chronic lymphocytic leukaemia in early phase. American Journal of Hematology 2017;92(6):E91-3. [DOI] [PubMed]
Barcelona‐Brno V ‐ Muñoz‐Novas 2018 (Spanish coh.) {published data only}
- Munoz-Novas C, Poza-Santaella M, Gonzalez-Gascon YMI, Hernandez-Sanchez M, Rodriguez-Vicente AE, Infante MS, et al. The international prognostic index for patients with chronic lymphocytic leukemia has the higher value in predicting overall outcome compared with the Barcelona-Brno biomarkers only prognostic model and the MD Anderson Cancer Center Prognostic Index. Biomed Research International 2018:9506979. [DOI: 10.1155/2018/9506979] [DOI] [PMC free article] [PubMed]
Barcelona‐Brno V ‐ Rani 2018 (Indian cohort) {published data only}
- Rani L, Gogia A, Singh V, Kumar L, Sharma A, Kaur G, et al. Comparative assessment of prognostic models in chronic lymphocytic leukemia: evaluation in Indian cohort. Annals of Hematology 2018;98(2):437-43. [DOI] [PubMed] [Google Scholar]
Barcelona‐Brno V ‐ Reda 2017 (Milan cohort) {published data only}
- Reda G, Cassin R, Fattizzo B, Giannarelli D, Mattiello V, Barcellini W, et al. Chronic lymphocytic leukemia and prognostic models: a bridge between clinical and biological markers. American Journal of Hematology 2017;92(7):E135-7. [DOI] [PubMed] [Google Scholar]
CLL‐IPI D ‐ Bahlo 2016 (development cohort) {published data only}
- International CLL-IPI working group. An international prognostic index for patients with chronic lymphocytic leukaemia (CLL-IPI): a meta-analysis of individual patient data. Lancet Oncology 2016;17(6):779-90. [DOI] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Bahlo 2016 (Mayo clinic 2001‐2014) {published data only}
- Gentile M, Shanafelt TD, Rossi D, Laurenti L, Mauro FR, Molica S, et al. Validation of the CLL-IPI and comparison with the MDACC prognostic index in newly diagnosed patients. Blood 2016;128(16):2093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *.International CLL-IPI Working Group. An international prognostic index for patients with chronic lymphocytic leukaemia (CLL-IPI): a meta-analysis of individual patient data. Lancet Oncology 2016;17(6):779-90. [DOI] [PubMed] [Google Scholar]
- Molica S, Shanafelt TD, Giannarelli D, Gentile M, Mirabelli R, Cutrona G, et al. The chronic lymphocytic leukemia international prognostic index predicts time to first treatment in early CLL: independent validation in a prospective cohort of early stage patients. American Journal of Hematology 2016;91:1090-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Bahlo 2016 (SCAN cohort) {published data only}
- International CLL-IPI working group. An international prognostic index for patients with chronic lymphocytic leukaemia (CLL-IPI): a meta-analysis of individual patient data. Lancet Oncology 2016;17(6):779-90. [DOI] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Da Cunha‐Bang 2016 (Danish cohort) {published data only}
- Da Cunha-Bang C, Christiansen I, Niemann CU. The CLL-IPI applied in a population-based cohort. Blood 2016;128(17):2181-3. [DOI] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Delgado 2017 (Barcelona cohort) {published data only}
- Delgado J, Doubek M, Baumann T, Kotaskova J, Molica S, Mozas P, et al. Chronic lymphocytic leukemia: a prognostic model comprising only two biomarkers (IGHV mutational status and FISH cytogenetics) separates patients with different outcome and simplifies the CLL-IPI. American Journal of Hematology 2017;92(4):375-80. [DOI] [PubMed]
CLL‐IPI V ‐ Gentile 2016 (Italian cohort) {published data only}
- Gentile M, Shanafelt TD, Mauro FR, Laurenti L, Rossi D, Molica S, et al. Comparison between the CLL-IPI and the Barcelona-Brno prognostic model: analysis of 1299 newly diagnosed cases. American Journal of Hematology 2017;93(2):E35-7. [DOI] [PubMed]
- *.Gentile M, Shanafelt TD, Rossi D, Laurenti L, Mauro FR, Molica S, et al. Validation of the CLL-IPI and comparison with the MDACC prognostic index in newly diagnosed patients. Blood 2016;128(16):2093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Molica 2016 (O‐CLL1‐GISL) {published data only}
- Molica S, Giannarelli D, Levato L, Mirabelli R, Gentile M, Morabito F. Assessing time to first treatment in early chronic lymphocytic leukemia (CLL): a comparative performance analysis of five prognostic models with inclusion of CLL-international prognostic index (CLL-IPI). Leukemia & Lymphoma 2017;58(7):1736-9. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Mirabelli R, Levato L, Gentile M, Morabito F, et al. Reliability of six prognostic models to predict time-to-first-treatment in patients with chronic lymphocytic leukaemia in early phase. American Journal of Hematology 2017;92(6):E91-3. [DOI] [PubMed]
- *.Molica S, Shanafelt TD, Giannarelli D, Gentile M, Mirabelli R, Cutrona G, et al. The chronic lymphocytic leukemia international prognostic index predicts time to first treatment in early CLL: independent validation in a prospective cohort of early stage patients. American Journal of Hematology 2016;91(11):1090-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Muñoz‐Novas 2018 (Spanish cohort) {published data only}
- Munoz-Novas C, Poza-Santaella M, Gonzalez-Gascon YMI, Hernandez-Sanchez M, Rodriguez-Vicente AE, Infante MS, et al. The international prognostic index for patients with chronic lymphocytic leukemia has the higher value in predicting overall outcome compared with the Barcelona-Brno biomarkers only prognostic model and the MD Anderson Cancer Center Prognostic Index. Biomed Research International 2018:9506979. [DOI: 10.1155/2018/9506979] [DOI] [PMC free article] [PubMed]
CLL‐IPI V ‐ Rani 2018 (Indian cohort) {published data only}
- Rani L, Gogia A, Singh V, Kumar L, Sharma A, Kaur G, et al. Comparative assessment of prognostic models in chronic lymphocytic leukemia: evaluation in Indian cohort. Annals of Hematology 2018;98(2):437-43. [DOI] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Reda 2017 (Milano cohort) {published data only}
- Reda G, Cassin R, Fattizzo B, Giannarelli D, Mattiello V, Barcellini W, et al. Chronic lymphocytic leukemia and prognostic models: a bridge between clinical and biological markers. American Journal of Hematology 2017;92(7):E135-7. [DOI] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Rigolin 2017 (Ferrera cohort) {published data only}
- Rigolin GM, Cavallari M, Quaglia FM, Formigaro L, Lista E, Urso A, et al. In CLL, comorbidities and the complex karyotype are associated with an inferior outcome independently of CLL-IPI. Blood 2017;129(26):3495-8. [DOI] [PubMed] [Google Scholar]
CLL‐IPI V ‐ Zhu 2018 (Chinese cohort) {published data only}
- Zhu HY, Wang L, Qiao J, Zou YX, Xia Y, Wu W, et al. Prognostic significance of CLL-IPI for Chinese patients with chronic lymphocytic leukemia. Chung Hua Hsueh Yeh Hsueh Tsa Chi 2018;39:392-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
GCLLSG D ‐ Pflug 2014 (GCLLSG) {published data only}
- *.Pflug N, Bahlo J, Shanafelt TD, Eichhorst BF, Bergmann MA, Elter T, et al. Development of a comprehensive prognostic index for patients with chronic lymphocytic leukemia. Blood 2014;124(1):49-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tam CS, Seymour JF. A new prognostic score for CLL. Blood 2014;124(1):1-2. [DOI] [PubMed] [Google Scholar]
GCLLSG V ‐ Molica 2015 (O‐CLL1‐GISL) {published data only}
- Molica S, Giannarelli D, Gentile M, Cutrona G, Di Renzo N, Di Raimondo F, et al. The utility of two prognostic models for predicting time to first treatment in early chronic lymphocytic leukemia patients: results of a comparative analysis. Leukemia Research 2013;37(8):943-7. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Levato L, Mirabelli R, Gentile M, Morabito F. Assessing time to first treatment in early chronic lymphocytic leukemia (CLL): a comparative performance analysis of five prognostic models with inclusion of CLL-international prognostic index (CLL-IPI). Leukemia & Lymphoma 2017;58(7):1736-9. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Mirabelli R, Levato L, Gentile M, Morabito F, et al. Reliability of six prognostic models to predict time-to-first-treatment in patients with chronic lymphocytic leukaemia in early phase. American Journal of Hematology 2017;92(6):E91-3. [DOI] [PubMed] [Google Scholar]
- *.Molica S, Giannarelli D, Mirabelli R, Levato L, Russo A, Linardi M, et al. Unavailability of thymidine kinase does not preclude the use of German comprehensive prognostic index: results of an external validation analysis in early chronic lymphocytic leukemia and comparison with MD Anderson Cancer Center model. European Journal of Haematology 2015;96(1):72-7. [DOI] [PubMed] [Google Scholar]
GCLLSG V ‐ Pflug 2014 (Mayo cohort) {published data only}
- Pflug N, Bahlo J, Shanafelt TD, Eichhorst BF, Bergmann MA, Elter T, et al. Development of a comprehensive prognostic index for patients with chronic lymphocytic leukemia. Blood 2014;124(1):49-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
GCLLSG V ‐ Rani 2018 (Indian cohort) {published data only}
- Rani L, Gogia A, Singh V, Kumar L, Sharma A, Kaur G, et al. Comparative assessment of prognostic models in chronic lymphocytic leukemia: evaluation in Indian cohort. Annals of Hematology 2018;98(2):437-43. [DOI] [PubMed] [Google Scholar]
GIMEMA D ‐ Molica 2005 (GIMEMA cohort) {published data only}
- Molica S, Mauro FR, Callea V, Gentile M, Giannarelli D, Lopez M, et al. A gender-based score system predicts the clinical outcome of patients with early B-cell chronic lymphocytic leukemia. Leukemia & Lymphoma 2005;46(4):553-60. [DOI] [PubMed] [Google Scholar]
GIMEMA V ‐ González Rodríguez 2009 (Cabueñes coh.) {published data only}
- Gonzalez Rodriguez AP, Gonzalez Garcia E, Fernandez Alvarez C, Gonzalez Huerta AJ, Gonzalez Rodriguez S. B-chronic lymphocytic leukemia: epidemiological study and comparison of MDACC and GIMENA prognostic indexes [Estudio epidemiológico y comparación de los índices pronósticos del MD Anderson Cancer Center y el índice del Gruppo Italiano Malattie Ematologiche Maligne dell' Adulto en pacientes con leucemia linfática crónica de células B]. Medicina Clinica 2009;133(5):161-6. [DOI] [PubMed] [Google Scholar]
MDACC 2007 D ‐ Wierda 2007 (MDACC) {published data only}
- Wierda WG, O'Brien S, Wang X, Faderl S, Ferrajoli A, Do KA, et al. Prognostic nomogram and index for overall survival in previously untreated patients with chronic lymphocytic leukemia. Blood 2007;109(11):4679-85. [DOI] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Bulian 2011 (Italian‐Swiss) {published data only}
- Bulian P, Tarnani M, Rossi D, Forconi F, Del Poeta G, Bertoni F, et al. Multicentre validation of a prognostic index for overall survival in chronic lymphocytic leukaemia. Hematological Oncology 2011;29(2):91-9. [DOI] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Gentile 2014 (Italian cohort) {published data only}
- *.Gentile M, Mauro FR, Rossi D, Vincelli I, Tripepi G, Recchia AG, et al. Italian external and multicentric validation of the MD Anderson Cancer Center nomogram and prognostic index for chronic lymphocytic leukaemia patients: analysis of 1502 cases. British Journal of Haematology 2014;167:224-32. [DOI] [PubMed] [Google Scholar]
- Gentile M, Shanafelt TD, Rossi D, Laurenti L, Mauro FR, Molica S, et al. Validation of the CLL-IPI and comparison with the MDACC prognostic index in newly diagnosed patients. Blood 2016;128(16):2093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Gentile 2016 (Mayo cohort) {published data only}
- Gentile M, Shanafelt TD, Cutrona G, Molica S, Tripepi G, Alvarez I, et al. A progression-risk score to predict treatment-free survival for early stage chronic lymphocytic leukemia patients. Leukemia 2016;30(6):1440-3. [DOI] [PubMed] [Google Scholar]
- *.Gentile M, Shanafelt TD, Rossi D, Laurenti L, Mauro FR, Molica S, et al. Validation of the CLL-IPI and comparison with the MDACC prognostic index in newly diagnosed patients. Blood 2016;128(16):2093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shanafelt TD, Jenkins G, Call TG, Zent CS, Slager S, Bowen DA, et al. Validation of a new prognostic index for patients with chronic lymphocytic leukemia. Cancer 2009;115:363-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
MDACC 2007 V ‐ González Rodríguez (Cabueñes) {published data only}
- Gonzalez Rodriguez AP, Gonzalez Garcia E, Fernandez Alvarez C, Gonzalez Huerta AJ, Gonzalez Rodriguez S. B-chronic lymphocytic leukemia: epidemiological study and comparison of MDACC and GIMENA prognostic indexes [Estudio epidemiológico y comparación de los índices pronósticos del MD Anderson Cancer Center y el índice del Gruppo Italiano Malattie Ematologiche Maligne dell' Adulto en pacientes con leucemia linfática crónica de células B]. Medicina Clinica 2009;133(5):161-6. [DOI] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Molica 2010 (GIMEMA cohort) {published data only}
- Molica S, Mauro FR, Callea V, Giannarelli D, Lauria F, Rotoli B, et al. The utility of a prognostic index for predicting time to first treatment in early chronic lymphocytic leukemia: the GIMEMA experience. Haematologica 2010;95(3):464-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Molica 2015 (O‐CLL1‐GISL) {published data only}
- Gentile M, Shanafelt TD, Cutrona G, Molica S, Tripepi G, Alvarez I, et al. A progression-risk score to predict treatment-free survival for early stage chronic lymphocytic leukemia patients. Leukemia 2016;30(6):1440-3. [DOI] [PubMed] [Google Scholar]
- Molica S, Di Raimondo F, Cutrona G, Fabris S, Mauro F, Brugiatelli M, et al. Clinical categories identified by a new prognostic index reflect biological characteristics of patients in early chronic lymphocytic leukemia: the Gruppo Italiano Studio Linfomi (GISL) experience. Leukemia Research 2010;34(8):e217-8. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Levato L, Mirabelli R, Gentile M, Morabito F. Assessing time to first treatment in early chronic lymphocytic leukemia (CLL): a comparative performance analysis of five prognostic models with inclusion of CLL-international prognostic index (CLL-IPI). Leukemia & Lymphoma 2017;58(7):1736-9. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Mirabelli R, Levato L, Gentile M, Morabito F, et al. Reliability of six prognostic models to predict time-to-first-treatment in patients with chronic lymphocytic leukaemia in early phase. American Journal of Hematology 2017;92(6):E91-3. [DOI] [PubMed] [Google Scholar]
- *.Molica S, Giannarelli D, Mirabelli R, Levato L, Russo A, Linardi M, et al. Unavailability of thymidine kinase does not preclude the use of German comprehensive prognostic index: results of an external validation analysis in early chronic lymphocytic leukemia and comparison with MD Anderson Cancer Center model. European Journal of Haematology 2015;96(1):72-7. [DOI] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Muñoz‐Novas 2018 (Spanish cohort) {published data only}
- Munoz-Novas C, Poza-Santaella M, Gonzalez-Gascon YMI, Hernandez-Sanchez M, Rodriguez-Vicente AE, Infante MS, et al. The international prognostic index for patients with chronic lymphocytic leukemia has the higher value in predicting overall outcome compared with the Barcelona-Brno biomarkers only prognostic model and the MD Anderson Cancer Center Prognostic Index. Biomed Research International 2018:9506979. [DOI: 10.1155/2018/9506979] [DOI] [PMC free article] [PubMed]
MDACC 2007 V ‐ Pflug 2014 (3 RCTs) {published data only}
- Pflug N, Bahlo J, Shanafelt TD, Eichhorst BF, Bergmann MA, Elter T, et al. Development of a comprehensive prognostic index for patients with chronic lymphocytic leukemia. Blood 2014;124(1):49-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Rani 2018 (Indian cohort) {published data only}
- Rani L, Gogia A, Singh V, Kumar L, Sharma A, Kaur G, et al. Comparative assessment of prognostic models in chronic lymphocytic leukemia: evaluation in Indian cohort. Annals of Hematology 2018;98(2):437-43. [DOI] [PubMed] [Google Scholar]
MDACC 2007 V ‐ Trajkova 2013 (Macedonia) {published data only}
- Trajkova S, Cevreska L, Pivkova-Veljanovska A, Ivanovski M, Dukovski D, Popova-Simjanovska M, et al. Multivariable model consisting of clinical and biological markers for time to first treatment in CLL patients: preliminary results from single centre experience. Makedonska Akademija na Naukite i Umetnostite Oddelenie Za Bioloshki i Meditsinski Nauki Prilozi 2013;34(3):39-48. [PubMed] [Google Scholar]
MDACC 2011 D ‐ Wierda 2011 (MDACC) {published data only}
- Wierda WG, O'Brien S, Wang X, Faderl S, Ferrajoli A, Do KA, et al. Multivariable model for time to first treatment in patients with chronic lymphocytic leukemia. Journal of Clinical Oncology 2011;29(31):4088-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
MDACC 2011 V ‐ Molica 2016 (O‐CLL1‐GISL) {published data only}
- Molica S, Giannarelli D, Gentile M, Cutrona G, Di Renzo N, Di Raimondo F, et al. External validation on a prospective basis of a nomogram for predicting the time to first treatment in patients with chronic lymphocytic leukemia. Cancer 2013;119(6):1177-85. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Gentile M, Cutrona G, Di Renzo N, Di Raimondo F, et al. The utility of two prognostic models for predicting time to first treatment in early chronic lymphocytic leukemia patients: results of a comparative analysis. Leukemia Research 2013;37(8):943-7. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Levato L, Mirabelli R, Gentile M, Lentini M, et al. A prognostic algorithm including a modified version of MD Anderson Cancer Center (MDACC) score predicts time to first treatment of patients with clinical monoclonal lymphocytosis (cMBL)/Rai stage 0 chronic lymphocytic leukemia (CLL). International Journal of Hematology 2014;100(3):290-5. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Levato L, Mirabelli R, Gentile M, Morabito F. Assessing time to first treatment in early chronic lymphocytic leukemia (CLL): a comparative performance analysis of five prognostic models with inclusion of CLL-international prognostic index (CLL-IPI). Leukemia & Lymphoma 2017;58(7):1736-9. [DOI] [PubMed] [Google Scholar]
- *.Molica S, Giannarelli D, Mirabelli R, Levato L, Gentile M, Morabito F, et al. Reliability of six prognostic models to predict time-to-first-treatment in patients with chronic lymphocytic leukaemia in early phase. American Journal of Hematology 2017;92(6):E91-3. [DOI] [PubMed] [Google Scholar]
- Molica S, Shanafelt TD, Giannarelli D, Gentile M, Mirabelli R, Cutrona G, et al. The chronic lymphocytic leukemia international prognostic index predicts time to first treatment in early CLL: independent validation in a prospective cohort of early stage patients. American Journal of Hematology 2016;91(11):1090-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
Morabito D ‐ Morabito 2009 (Italian cohort) {published data only}
- Morabito F, Cutrona G, Gentile M, Matis S, Todoerti K, Colombo M, et al. Definition of progression risk based on combinations of cellular and molecular markers in patients with Binet stage A chronic lymphocytic leukaemia. British Journal of Haematology 2009;146(1):44-53. [DOI] [PubMed] [Google Scholar]
Morabito V ‐ Gentile 2014 (O‐CLL1‐GISL) {published data only}
- Gentile M, Cutrona G, Mosca L, Matis S, Fabris S, Lionetti M, et al. Prospective validation of a risk score based on biological markers for predicting progression free survival in Binet stage A chronic lymphocytic leukemia patients: results of the multicenter O-CLL1-GISL study. American Journal of Hematology 2014;89(7):743-50. [DOI] [PubMed] [Google Scholar]
O‐CLL‐1 D ‐ Gentile 2016 (O‐CLL‐1‐GISL) {published data only}
- *.Gentile M, Shanafelt TD, Cutrona G, Molica S, Tripepi G, Alvarez I, et al. A progression-risk score to predict treatment-free survival for early stage chronic lymphocytic leukemia patients. Leukemia 2016;30(6):1440-3. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Levato L, Mirabelli R, Gentile M, Morabito F. Assessing time to first treatment in early chronic lymphocytic leukemia (CLL): a comparative performance analysis of five prognostic models with inclusion of CLL-international prognostic index (CLL-IPI). Leukemia & Lymphoma 2017;58(7):1736-9. [DOI] [PubMed] [Google Scholar]
- Molica S, Giannarelli D, Mirabelli R, Levato L, Gentile M, Morabito F, et al. Reliability of six prognostic models to predict time-to-first-treatment in patients with chronic lymphocytic leukaemia in early phase. American Journal of Hematology 2017;92(6):E91-3. [DOI] [PubMed] [Google Scholar]
- Molica S, Shanafelt TD, Giannarelli D, Gentile M, Mirabelli R, Cutrona G, et al. The chronic lymphocytic leukemia international prognostic index predicts time to first treatment in early CLL: independent validation in a prospective cohort of early stage patients. American Journal of Hematology 2016;91(11):1090-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
O‐CLL1 V ‐ Gentile 2016 (Mayo cohort) {published data only}
- Gentile M, Shanafelt TD, Cutrona G, Molica S, Tripepi G, Alvarez I, et al. A progression-risk score to predict treatment-free survival for early stage chronic lymphocytic leukemia patients. Leukemia 2016;30(6):1440-3. [DOI] [PubMed] [Google Scholar]
O‐CLL‐1 V ‐ Rani 2018 (Indian cohort) {published data only}
- Rani L, Gogia A, Singh V, Kumar L, Sharma A, Kaur G, et al. Comparative assessment of prognostic models in chronic lymphocytic leukemia: evaluation in Indian cohort. Annals of Hematology 2018;98(2):437-43. [DOI] [PubMed] [Google Scholar]
Rossi D ‐ Rossi 2013 (Italian cohort) {published data only}
- Rossi D, Rasi S, Spina V, Bruscaggin A, Monti S, Ciardullo C, et al. Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia. Blood 2013;121(8):1403-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
Rossi V ‐ Jeromin 2014 (Munich cohort) {published data only}
- Jeromin S, Weissmann S, Haferlach C, Dicker F, Bayer K, Grossmann V, et al. SF3B1 mutations correlated to cytogenetics and mutations in NOTCH1, FBXW7, MYD88, XPO1 and TP53 in 1160 untreated CLL patients. Leukemia 2014;28(1):108-17. [DOI] [PubMed] [Google Scholar]
Rossi V ‐ Rossi 2013 (unclear) {published data only}
- Rossi D, Rasi S, Spina V, Bruscaggin A, Monti S, Ciardullo C, et al. Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia. Blood 2013;121(8):1403-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
Stephens OS D ‐ Stephens 2015 (Ohio cohort) {published data only}
- Stephens DM, Ruppert AS, Weirda WG, Jones JA, Woyach JA, Maddocks K, et al. Externally validated predictive clinical model for untreated del(17p13.1) chronic lymphocytic leukemia patients. American Journal of Hematology 2015;90(11):967-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Stephens OS V ‐ Stephens 2015 (MDACC) {published data only}
- Stephens DM, Ruppert AS, Weirda WG, Jones JA, Woyach JA, Maddocks K, et al. Externally validated predictive clinical model for untreated del(17p13.1) chronic lymphocytic leukemia patients. American Journal of Hematology 2015;90(11):967-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Stephens TFS D ‐ Stephens 2015 (Ohio cohort) {published data only}
- Stephens DM, Ruppert AS, Weirda WG, Jones JA, Woyach JA, Maddocks K, et al. Externally validated predictive clinical model for untreated del(17p13.1) chronic lymphocytic leukemia patients. American Journal of Hematology 2015;90(11):967-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Stephens TFS V ‐ Stephens 2015 (MDACC) {published data only}
- Stephens DM, Ruppert AS, Weirda WG, Jones JA, Woyach JA, Maddocks K, et al. Externally validated predictive clinical model for untreated del(17p13.1) chronic lymphocytic leukemia patients.. American Journal of Hematology 2015;90(11):967-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
References to studies excluded from this review
Apelgren 2006 {published data only}
- Apelgren P, Hasselblom S, Werlenius O, Nilsson-Ehle H, Andersson PO. Evaluation of clinical staging in chronic lymphocytic leukemia - population-based study. Leukemia & Lymphoma 2006;47(12):2505-16. [DOI] [PubMed] [Google Scholar]
Baccarani 1982 {published data only}
- Baccarani M, Cavo M, Gobbi M, Lauria F, Tura S. Staging of chronic lymphocytic leukemia. Blood 1982;59(6):1191-6. [PubMed] [Google Scholar]
Baliakas 2015 {published data only}
- Baliakas P, Agathangelidis A, Hadzidimitriou A, Sutton LA, Minga E, Tsanousa A, et al. Not all IGHV3-21 chronic lymphocytic leukemias are equal: prognostic considerations. Blood 2015;125(5):856-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Berke 2019 {published data only}
- Berke Mentese I, Yegin ZA, Gokcen S, Ozkurt ZN, Yagci M. Prognostic significance of serum BAFF, APRIL, TACI and BCMA levels in chronic lymphocytic leukemia. Indian Journal of Hematology and Blood Transfusion 2019;35(2):265-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
Bettini 1986 {published data only}
- Bettini R, Rapazzini P, Ferrari V, Steidl L. Clinical staging of chronic lymphatic leukemia. Recenti Progressi in Medicina 1986;77(2):72-6. [PubMed] [Google Scholar]
Binet 1977 {published data only}
- Binet JL, Leporrier M, Dighiero G, Charron D, D'Athis P, Vaugier G, et al. A clinical staging system for chronic lymphocytic leukemia: prognostic significance. Cancer 1977;40(2):855-64. [DOI] [PubMed] [Google Scholar]
Binet 1981 {published data only}
Bo 2014 {published data only}
- Bo MD, Del Principe MI, Pozzo F, Ragusa D, Bulian P, Rossi D, et al. NOTCH1 mutations identify a chronic lymphocytic leukemia patient subset with worse prognosis in the setting of a rituximab-based induction and consolidation treatment. Annals of Hematology 2014;93(10):1765-74. [DOI] [PubMed] [Google Scholar]
Bomben 2005 {published data only}
- Bomben R, Dal Bo M, Zucchetto A, Zaina E, Nanni P, Sonego P, et al. Mutational status of IgV(H) genes in B-cell chronic lymphocytic leukemia and prognosis: percent mutations or antigen-driven selection? Leukemia 2005;19(8):1490-2. [DOI] [PubMed] [Google Scholar]
Bomben 2009 {published data only}
- Bomben R, Dal Bo M, Capello D, Forconi F, Maffei R, Laurenti L, et al. Molecular and clinical features of chronic lymphocytic leukaemia with stereotyped B cell receptors: results from an Italian multicentre study. British Journal of Haematology 2009;144(4):492-506. [DOI] [PubMed] [Google Scholar]
Bou Samra 2014 {published data only}
- Bou Samra E, Klein B, Commes T, Moreaux J. Identification of a 20-gene expression-based risk score as a predictor of clinical outcome in chronic lymphocytic leukemia patients. Biomed Research International 2014;2014:423174. [DOI] [PMC free article] [PubMed] [Google Scholar]
Brejcha 2010 {published data only}
- Brejcha M, Doubek M, Cmunt E, Schwarz J, Pospisil Z, Kozmon P, et al. New prognostic markers of chronic lymphocytic leukemia (CLL) in the everyday hematological practice. An analysis of data from four hematology departments. [Czech]. Transfuze a Hematologie Dnes 2010;16(Suppl 1):62-6. [Google Scholar]
Brugiatelli 2007 {published data only}
- Brugiatelli M, Loteta B, Nocilli L, Mannina D. B-cell chronic lymphocytic leukemia: clinical impact of biological prognostic factors and updated treatment strategies. Lijecnicki Vjesnik 2007;129 Suppl 3:26-8. [PubMed] [Google Scholar]
Bulian 2014 {published data only}
- Bulian P, Shanafelt TD, Fegan C, Zucchetto A, Cro L, Nuckel H, et al. CD49d is the strongest flow cytometry-based predictor of overall survival in chronic lymphocytic leukemia. Journal of Clinical Oncology 2014;32(9):897-904. [DOI] [PMC free article] [PubMed] [Google Scholar]
Byrd 2006 {published data only}
- Byrd JC, Gribben JG, Peterson BL, Grever MR, Lozanski G, Lucas DM, et al. Select high-risk genetic features predict earlier progression following chemoimmunotherapy with fludarabine and rituximab in chronic lymphocytic leukemia: justification for risk-adapted therapy. Journal of Clinical Oncology 2006;24(3):437-43. [DOI] [PubMed] [Google Scholar]
Cailliod 2005 {published data only}
- Cailliod R, Quantin C, Carli PM, Jooste V, Le Teuff G, Binquet C, et al. A population-based assessment of the prognostic value of the CD19 positive lymphocyte count in B-cell chronic lymphocytic leukemia using Cox and Markov models. European Journal of Epidemiology 2005;20(12):993-1001. [DOI] [PubMed] [Google Scholar]
Callea 1999 {published data only}
- Callea V, Morabito F, Oliva BM, Stelitano C, Levato D, Dattilo A, et al. Surface CD14 positivity in B-cell chronic lymphocytic leukaemia is related to clinical outcome. British Journal of Haematology 1999;107(2):347-52. [DOI] [PubMed] [Google Scholar]
Catovsky 1989 {published data only}
- Catovsky D, Fooks J, Richards S, MRC Working Party on Leukaemia in Adults. Prognostic factors in chronic lymphocytic leukaemia: the importance of age, sex and response to treatment in survival. A report from the MRC CLL 1 trial. British Journal of Haematology 1989;72(2):141-9. [DOI] [PubMed] [Google Scholar]
Cesano 2013 {published data only}
- Cesano A, Perbellini O, Evensen E, Chu CC, Cioffi F, Ptacek J, et al. Association between B-cell receptor responsiveness and disease progression in B-cell chronic lymphocytic leukemia: results from single cell network profiling studies. Haematologica 2013;98(4):626-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
Chang 2003 {published data only}
- Chang CC, Liu CZ, Cleveland RP. Relative importance of CD38 expression over myeloid-associated markers expression in predicting the clinical course of B-CLL patients. Leukemia and Lymphoma 2003;44(6):977-82. [DOI] [PubMed] [Google Scholar]
Chastang 1985 {published data only}
- Chastang C, Travade P, Auquier A. Critical discussion of the assessment of a three-stage prognostic classification for chronic lymphocytic leukemia. Statistics in Medicine 1985;4(3):287-93. [DOI] [PubMed] [Google Scholar]
Chauzeix 2018 {published data only}
- Chauzeix J, Laforet MP, Deveza M, Crowther L, Marcellaud E, Derouault P, et al. Normal serum protein electrophoresis and mutated IGHV genes detect very slowly evolving chronic lymphocytic leukemia patients. Cancer Medicine 2018;7(6):2621–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Chelazzi 1979 {published data only}
- Chelazzi G, Bettini R, Michetti A, Ricci G. Prognostic value of the clinical staging of chronic lymphocytic leukemia (author's transl). Haematologica 1979;64(4):463-71. [PubMed] [Google Scholar]
Chen 1997 {published data only}
- Chen PM, Lin SH, Fan SF, Chiou TJ, Hsieh RK, Yu IT, et al. Genotypic characterization and multivariate survival analysis of chronic lymphocytic leukemia in Taiwan. Acta Haematologica 1997;97(4):196-204. [DOI] [PubMed] [Google Scholar]
Chena 2008 {published data only}
- Chena C, Avalos JS, Bezares RF, Arrossagaray G, Turdo K, Bistmans A, et al. Biallelic deletion 13q14.3 in patients with chronic lymphocytic leukemia: cytogenetic, FISH and clinical studies. European Journal of Haematology 2008;81(2):94-9. [DOI] [PubMed] [Google Scholar]
Chevallier 2002 {published data only}
- Chevallier P, Penther D, Avet-Loiseau H, Robillard N, Ifrah N, Mahe B, et al. CD38 expression and secondary 17p deletion are important prognostic factors in chronic lymphocytic leukaemia. British Journal of Haematology 2002;116(1):142-50. [DOI] [PubMed] [Google Scholar]
Chiaretti 2014 {published data only}
- Chiaretti S, Marinelli M, Del Giudice I, Bonina S, Piciocchi A, Messina M, et al. NOTCH1, SF3B1, BIRC3 and TP53 mutations in patients with chronic lymphocytic leukemia undergoing first-line treatment: correlation with biological parameters and response to treatment. Leukemia & Lymphoma 2014;55(12):2785-92. [DOI] [PubMed] [Google Scholar]
Christiansen 1994 {published data only}
- Christiansen I, Gidlof C, Wallgren AC, Simonsson B, Totterman TH. Serum levels of soluble intercellular adhesion molecule 1 are increased in chronic B-lymphocytic leukemia and correlate with clinical stage and prognostic markers. Blood 1994;84(9):3010-6. [PubMed] [Google Scholar]
Chuang 2012 {published data only}
- Chuang HY, Rassenti L, Salcedo M, Licon K, Kohlmann A, Haferlach T, et al. Subnetwork-based analysis of chronic lymphocytic leukemia identifies pathways that associate with disease progression. Blood 2012;120(13):2639-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
Ciccone 2012 {published data only}
- Ciccone M, Agostinelli C, Rigolin GM, Piccaluga PP, Cavazzini F, Righi S, et al. Proliferation centers in chronic lymphocytic leukemia: correlation with cytogenetic and clinicobiological features in consecutive patients analyzed on tissue microarrays. Leukemia 2012;26(3):499-508. [DOI] [PubMed] [Google Scholar]
Ciocoiu 1988 {published data only}
- Ciocoiu A, Motoiu I, Niculescu R, Berceanu S. The staging of chronic lymphatic leukemia. Its prognostic value and therapeutic implications. Revista de Medicină Internă, Neurologe, Psihiatrie, Neurochirurgie, Dermato-venerologie 1988;40(1):41-5. [PubMed] [Google Scholar]
Claus 2012 {published data only}
- Claus R, Lucas DM, Stilgenbauer S, Ruppert AS, Yu L, Zucknick M, et al. Quantitative DNA methylation analysis identifies a single CpG dinucleotide important for ZAP-70 expression and predictive of prognosis in chronic lymphocytic leukemia. Journal of Clinical Oncology 2012;30(20):2483-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
Claus 2014 {published data only}
- Claus R, Lucas DM, Ruppert AS, Williams KE, Weng D, Patterson K, et al. Validation of ZAP-70 methylation and its relative significance in predicting outcome in chronic lymphocytic leukemia. Blood 2014;124(1):42-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Cmunt 2002 {published data only}
- Cmunt E, Michalova K, Sindelarova L, Karban J, Zemanova Z, Kurkova S, et al. Importance of prognostic factors in patients with chronic B-lymphocytic leukemia at the time of diagnosis. Sbornik Lekarsky 2002;103(3):359-70. [PubMed] [Google Scholar]
Cocco 2005 {published data only}
- Cocco AE, Osei ES, Thut DM, Edinger AK, Powers JJ, Fu P, et al. Bimodal cell populations are common in chronic lymphocytic leukemia but do not impact overall survival. American Journal of Clinical Pathology 2005;123(6):818-25. [DOI] [PubMed] [Google Scholar]
Corcoran 2005 {published data only}
- Corcoran M, Parker A, Orchard J, Davis Z, Wirtz M, Schmitz OJ, et al. ZAP-70 methylation status is associated with ZAP-70 expression status in chronic lymphocytic leukemia. Haematologica 2005;90(8):1078-88. [PubMed] [Google Scholar]
Cordone 1998 {published data only}
- Cordone I, Masi S, Mauro FR, Soddu S, Morsilli O, Valentini T, et al. p53 expression in B-cell chronic lymphocytic leukemia: a marker of disease progression and poor prognosis. Blood 1998;91(11):4342-9. [PubMed] [Google Scholar]
Cortese 2014 {published data only}
- Cortese D, Sutton LA, Cahill N, Smedby KE, Geisler C, Gunnarsson R, et al. On the way towards a 'CLL prognostic index': focus on TP53, BIRC3, SF3B1, NOTCH1 and MYD88 in a population-based cohort. Leukemia 2014;28(3):710-3. [DOI] [PubMed] [Google Scholar]
Coscia 2012 {published data only}
- Coscia M, Vitale C, Peola S, Foglietta M, Rigoni M, Griggio V, et al. Dysfunctional V9V2 T cells are negative prognosticators and markers of dysregulated mevalonate pathway activity in chronic lymphocytic leukemia cells. Blood 2012;120(16):3271-9. [DOI] [PubMed] [Google Scholar]
Crespo 2003 {published data only}
- Crespo M, Bosch F, Villamor N, Bellosillo B, Colomer D, Rozman M, et al. ZAP-70 expression as a surrogate for immunoglobulin-variable-region mutations in chronic lymphocytic leukemia. New England Journal of Medicine 2003;348(18):1764-75. [DOI] [PubMed] [Google Scholar]
Criel 1997 {published data only}
- Criel A, Verhoef G, Vlietinck R, Mecucci C, Billiet J, Michaux L, et al. Further characterization of morphologically defined typical and atypical CLL: a clinical, immunophenotypic, cytogenetic and prognostic study on 390 cases. British Journal of Haematology 1997;97(2):383-91. [DOI] [PubMed] [Google Scholar]
Cro 2009 {published data only}
- Cro L, Morabito F, Zucal N, Fabris S, Lionetti M, Cutrona G, et al. CD26 expression in mature B-cell neoplasia: its possible role as a new prognostic marker in B-CLL. Hematological Oncology 2009;27(3):140-7. [DOI] [PubMed] [Google Scholar]
Cro 2010 {published data only}
- Cro L, Ferrario A, Lionetti M, Bertoni F, Zucal NN, Nobili L, et al. The clinical and biological features of a series of immunophenotypic variant of B-CLL. European Journal of Haematology 2010;85(2):120-9. [DOI] [PubMed] [Google Scholar]
Cuneo 2004 {published data only}
- Cuneo A, Rigolin GM, Bigoni R, De Angeli C, Veronese A, Cavazzini F, et al. Chronic lymphocytic leukemia with 6q- shows distinct hematological features and intermediate prognosis. Leukemia 2004;18(3):476-83. [DOI] [PubMed] [Google Scholar]
Cuneo 2018 {published data only}
- Cuneo A, Follows G, Rigolin GM, Piciocchi A, Tedeschi A, Trentin L, et al. Efficacy of bendamustine and rituximab as first salvage treatment in chronic lymphocytic leukemia and indirect comparison with ibrutinib: a GIMEMA, ERIC and UK CLL FORUM study. Haematologica 2018;103(7):1209-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
D'Arena 2001 {published data only}
- D'Arena G, Musto P, Cascavilla N, Dell'Olio M, Di Renzo N, Perla G, et al. CD38 expression correlates with adverse biological features and predicts poor clinical outcome in B-cell chronic lymphocytic leukemia. Leukemia & Lymphoma 2001;42(1-2):109-14. [DOI] [PubMed] [Google Scholar]
D'Arena 2007 {published data only}
- D'Arena G, Tarnani M, Rumi C, Vaisitti T, Aydin S, De Filippi R, et al. Prognostic significance of combined analysis of ZAP-70 and CD38 in chronic lymphocytic leukemia. American Journal of Hematology 2007;82(9):787-91. [DOI] [PubMed] [Google Scholar]
D'Arena 2012 {published data only}
- D'Arena G, D'Auria F, Simeon V, Laurenti L, Deaglio S, Mansueto G, et al. A shorter time to the first treatment may be predicted by the absolute number of regulatory T-cells in patients with Rai stage 0 chronic lymphocytic leukemia. American Journal of Hematology 2012;87(6):628-31. [DOI] [PubMed] [Google Scholar]
Damle 1999 {published data only}
- Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 1999;94(6):1840-7. [PubMed] [Google Scholar]
Dasgupta 2015 {published data only}
- Dasgupta A, Mahapatra M, Saxena R. A study for proposal of use of regulatory T cells as a prognostic marker and establishing an optimal threshold level for their expression in chronic lymphocytic leukemia. Leukemia & Lymphoma 2015;56(6):1831-8. [DOI] [PubMed] [Google Scholar]
Davis 2016 {published data only}
- Davis Z, Forconi F, Parker A, Gardiner A, Thomas P, Catovsky D, et al. The outcome of Chronic lymphocytic leukaemia patients with 97% IGHV gene identity to germline is distinct from cases with < 97% identity and similar to those with 98% identity. British Journal of Haematology 2016;173(1):127-36. [DOI] [PubMed] [Google Scholar]
DeAndres‐Galiana 2016 {published data only}
- DeAndres-Galiana EJ, Fernandez-Martinez JL, Luaces O, Del Coz JJ, Huergo-Zapico L, Acebes-Huerta A, et al. Analysis of clinical prognostic variables for chronic lymphocytic leukemia decision-making problems. Journal of Biomedical Informatics 2016;60:342-51. [DOI] [PubMed] [Google Scholar]
De Faria 2000 {published data only}
- De Faria JR, De Oliveira JS, Delbone de Faria RM, Silva MR, Goihman S, Yamamoto M, et al. Prognosis related to staging systems for chronic lymphocytic leukemia. Revista Paulista de Medicina [Sao Paulo Medical Journal] 2000;118(4):83-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Degan 2004 {published data only}
- Degan M, Bomben R, Bo MD, Zucchetto A, Nanni P, Rupolo M, et al. Analysis of IgV gene mutations in B cell chronic lymphocytic leukaemia according to antigen-driven selection identifies subgroups with different prognosis and usage of the canonical somatic hypermutation machinery. British Journal of Haematology 2004;126(1):29-42. [DOI] [PubMed] [Google Scholar]
Delgado 2009 {published data only}
- Delgado J, Pratt G, Phillips N, Briones J, Fegan C, Nomdedeu J, et al. Beta2-microglobulin is a better predictor of treatment-free survival in patients with chronic lymphocytic leukaemia if adjusted according to glomerular filtration rate. British Journal of Haematology 2009;145(6):801-5. [DOI] [PubMed] [Google Scholar]
Delgado 2014 {published data only}
- Delgado J, Salaverria I, Baumann T, Martinez-Trillos A, Lee E, Jimenez L, et al. Genomic complexity and IGHV mutational status are key predictors of outcome of chronic lymphocytic leukemia patients with TP53 disruption. Haematologica 2014;99(11):e231-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
Del Guidice 2011 {published data only}
- Del Giudice I, Mauro FR, De Propris MS, Santangelo S, Marinelli M, Peragine N, et al. White blood cell count at diagnosis and immunoglobulin variable region gene mutations are independent predictors of treatment-free survival in young patients with stage A chronic lymphocytic leukemia. Haematologica 2011;96(4):626-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
Del Poeta 2010 {published data only}
- Del Poeta G, Del Principe MI, Maurillo L, Rossi FM, Buccisano F, Ammatuna E, et al. Spontaneous apoptosis and proliferation detected by BCL-2 and CD71 proteins are important progression indicators within ZAP-70 negative chronic lymphocytic leukemia. Leukemia & Lymphoma 2010;51(1):95-106. [DOI] [PubMed] [Google Scholar]
Del Principe 2004 {published data only}
- Del Principe MI, Del Poeta G, Venditti A, Buccisano F, Maurillo L, Marini R, et al. Clinical significance of soluble p53 protein in B-cell chronic lymphocytic leukemia. Haematologica 2004;89(12):1468-75. [PubMed] [Google Scholar]
Del Principe 2006 {published data only}
- Del Principe MI, Del Poeta G, Buccisano F, Maurillo L, Venditti A, Zucchetto A, et al. Clinical significance of ZAP-70 protein expression in B-cell chronic lymphocytic leukemia. Blood 2006;108(3):853-61. [DOI] [PubMed] [Google Scholar]
De Rossi 1989 {published data only}
- De Rossi G. Prognosis in chronic lymphocytic leukemia (CLL): experience of the Italian cooperative group (GIMEMA). Bone Marrow Transplantation 1989;4 Suppl 1:162-4. [PubMed] [Google Scholar]
Deslandes 2007 {published data only}
- Deslandes E, Chevret S. Assessing surrogacy from the joint modelling of multivariate longitudinal data and survival: application to clinical trial data on chronic lymphocytic leukaemia. Statistics in Medicine 2007;26(30):5411-21. [DOI] [PubMed] [Google Scholar]
Dimier 2018 {published data only}
- Dimier N, Delmar P, Ward C, Morariu-Zamfir R, Fingerle-Rowson G, Bahlo J, et al. A model for predicting effect of treatment on progression-free survival using MRD as a surrogate end point in CLL. Blood 2018;131(9):955-62. [DOI] [PubMed] [Google Scholar]
Di Raimondo 2001 {published data only}
- Di Raimondo F, Giustolisi R, Lerner S, Cacciola E, O'Brien S, Kantarjian H, et al. Retrospective study of the prognostic role of serum thymidine kinase level in CLL patients with active disease treated with fludarabine. Annals of Oncology 2001;12(5):621-5. [DOI] [PubMed] [Google Scholar]
Dong 2011 {published data only}
- Dong HJ, Zhou LT, Zhu DX, Wang DM, Fang C, Zhu HY, et al. The prognostic significance of TP53 mutations in Chinese patients with chronic lymphocytic leukemia is independent of del(17p13). Annals of Hematology 2011;90(6):709-17. [DOI] [PubMed] [Google Scholar]
Dong 2014 {published data only}
- Dong HJ, Fang C, Wang L, Fan L, Xu J, Wu JZ, et al. TP53 Pro72 allele potentially increases the poor prognostic significance of TP53 mutation in chronic lymphocytic leukemia. Medical Oncology 2014;31(4):908. [DOI] [PubMed] [Google Scholar]
Durak 2009 {published data only}
- Durak B, Akay OM, Aslan V, Ozdemir M, Sahin F, Artan S, et al. Prognostic impact of chromosome alterations detected by FISH in Turkish patients with B-cell chronic lymphocytic leukemia. Cancer Genetics and Cytogenetics 2009;188(2):65-9. [DOI] [PubMed] [Google Scholar]
El‐Kinawy 2012 {published data only}
- El-Kinawy NS, Sharaf HM, El-Hamid MA. Prognostic significance of del 17p, ZAP-70 and CD38 as independent indicators for B-CLL: correlation to response to treatment and disease outcome. Egyptian Journal of Medical Human Genetics 2012;13(2):173-81. [Google Scholar]
Fang 2019 {published data only}
- Fang H, Reichard KK, Rabe KG, Hanson CA, Call TG, Ding W, et al. IGH translocations in chronic lymphocytic leukemia: clinicopathologic features and clinical outcomes. American Journal of Hematology 2019;94(3):338-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
Ferrara 1981 {published data only}
- Ferrara F, Borrelli G, Fasanaro A, Mettivier V, Rametta V, Cimino R. Prognostic evaluation of chronic lymphocytic leukemia in relation to the Rai classification. Critical analysis of 145 personal cases. Minerva Medica 1981;72(12):741-4. [PubMed] [Google Scholar]
Ferreira 2014 {published data only}
- Ferreira PG, Jares P, Rico D, Gomez-Lopez G, Martinez-Trillos A, Villamor N, et al. Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia. Genome Research 2014;24(2):212-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
French Cooperative Group on CLL 1988 {published data only}
- French Cooperative Group on Chronic Lymphocytic Leukemia. Comparison of the (A, B, C) staging and the Rai's staging from a large prospective series (935 patients). Nouvelle Revue Française d'Hématologie 1988;30:363-7. [PubMed] [Google Scholar]
Friedman 2009 {published data only}
- Friedman DR, Weinberg JB, Barry WT, Goodman BK, Volkheimer AD, Bond KM, et al. A genomic approach to improve prognosis and predict therapeutic response in chronic lymphocytic leukemia. Clinical Cancer Research 2009;15(22):6947-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
Gattei 2008 {published data only}
- Gattei V, Bulian P, Del Principe MI, Zucchetto A, Maurillo L, Buccisano F, et al. Relevance of CD49d protein expression as overall survival and progressive disease prognosticator in chronic lymphocytic leukemia. Blood 2008;111(2):865-73. [DOI] [PubMed] [Google Scholar]
Gdynia 2018 {published data only}
- Gdynia G, Robak T, Kopitz J, Heller A, Grekova S, Duglova K, et al. Distinct activities of glycolytic enzymes identify chronic lymphocytic leukemia patients with a more aggressive course and resistance to chemo-immunotherapy. EBioMedicine 2018;32:125-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
Geisler 1997 {published data only}
- Geisler CH, Philip P, Christensen BE, Hou-Jensen K, Pedersen NT, Jensen OM, et al. In B-cell chronic lymphocytic leukaemia chromosome 17 abnormalities and not trisomy 12 are the single most important cytogenetic abnormalities for the prognosis: a cytogenetic and immunophenotypic study of 480 unselected newly diagnosed patients. Leukemia Research 1997;21(11-12):1011-23. [DOI] [PubMed] [Google Scholar]
Gentile 2016 {published data only}
- Gentile M, Cutrona G, Molica S, Ilariucci F, Mauro FR, Di Renzo N, et al. Prospective validation of predictive value of abdominal computed tomography scan on time to first treatment in Rai 0 chronic lymphocytic leukemia patients: results of the multicenter O-CLL1-GISL study. European Journal of Haematology 2016;96(1):36-45. [DOI] [PubMed] [Google Scholar]
Gentile 2018 {published data only}
- Gentile M, Shanafelt TD, Mauro FR, Reda G, Rossi D, Laurenti L, et al. Predictive value of the CLL-IPI in CLL patients receiving chemo-immunotherapy as first-line treatment. European Journal of Haematology 2018;101(5):703-6. [DOI] [PubMed] [Google Scholar]
Giles 2003 {published data only}
- Giles FJ, Bekele BN, O'Brien S, Cortes JE, Verstovsek S, Balerdi M, et al. A prognostic model for survival in chronic lymphocytic leukaemia based on p53 expression. British Journal of Haematology 2003;121(4):578-85. [DOI] [PubMed] [Google Scholar]
Giudice 2018 {published data only}
- Giudice ID, Rigolin GM, Raponi S, Cafforio L, Ilari C, Wang J, et al. Refined karyotype-based prognostic stratification of chronic lymphocytic leukemia with a low- and very-low-risk genetic profile. Leukemia 2018;32(2):543-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
Gogia 2014 {published data only}
- Gogia A, Raina V, Gupta R, Gajendra S, Kumar L, Sharma A, et al. Prognostic and predictive significance of smudge cell percentage on routine blood smear in chronic lymphocytic leukemia. Clinical Lymphoma, Myeloma & Leukemia 2014;14(6):514-7. [DOI] [PubMed] [Google Scholar]
Gonzalez 2013 {published data only}
- Gonzalez D, Else M, Wren D, Usai M, Buhl AM, Parker A, et al. CLLU1 expression has prognostic value in chronic lymphocytic leukemia after first-line therapy in younger patients and in those with mutated IGHV genes. Haematologica 2013;98(2):274-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Gonzalez‐Gascon 2015 {published data only}
- Gonzalez-Gascon YMI, Hernandez-Sanchez M, Rodriguez-Vicente AE, Sanzo C, Aventin A, Puiggros A, et al. A high proportion of cells carrying trisomy 12 is associated with a worse outcome in patients with chronic lymphocytic leukemia. Hematological Oncology 2015;17(10):84–92. [DOI] [PubMed] [Google Scholar]
Gonzalez‐Rodriguez 2010 {published data only}
- Gonzalez-Rodriguez AP, Contesti J, Huergo-Zapico L, Lopez-Soto A, Fernandez-Guizan A, Acebes-Huerta A, et al. Prognostic significance of CD8 and CD4 T cells in chronic lymphocytic leukemia. Leukemia & Lymphoma 2010;51(10):1829-36. [DOI] [PubMed] [Google Scholar]
Grabowski 2005 {published data only}
- Grabowski P, Hultdin M, Karlsson K, Tobin G, Aleskog A, Thunberg U, et al. Telomere length as a prognostic parameter in chronic lymphocytic leukemia with special reference to VH gene mutation status. Blood 2005;105(12):4807-12. [DOI] [PubMed] [Google Scholar]
Grever 2006 {published data only}
- Grever MR, Dewald GW, Neuberg DS, Reed JC, Kitada S, Flinn IW, et al. Select high risk genetic features predict earlier progression following chemotherapy in chronic lymphocytic leukemia: prospective randomized trial (Intergroup E2997) to evaluate justification for risk-adapted therapy [abstract]. Journal of Clinical Oncology: ASCO Annual Meeting Proceedings 2006;24:342. [Google Scholar]
Hallek 1999 {published data only}
- Hallek M, Langenmayer I, Nerl C, Knauf W, Dietzfelbinger H, Adorf D, et al. Elevated serum thymidine kinase levels identify a subgroup at high risk of disease progression in early, nonsmoldering chronic lymphocytic leukemia. Blood 1999;93(5):1732-7. [PubMed] [Google Scholar]
Han 1984 {published data only}
- Han T, Barcos M, Emrich L, Ozer H, Gajera R, Gomez GA, et al. Bone marrow infiltration patterns and their prognostic significance in chronic lymphocytic leukemia: correlations with clinical, immunologic, phenotypic, and cytogenetic data. Journal of Clinical Oncology 1984;2(6):562-70. [DOI] [PubMed] [Google Scholar]
Herold 2011 {published data only}
- Herold T, Jurinovic V, Metzeler KH, Boulesteix AL, Bergmann M, Seiler T, et al. An eight-gene expression signature for the prediction of survival and time to treatment in chronic lymphocytic leukemia. Leukemia 2011;25(10):1639-45. [DOI] [PubMed] [Google Scholar]
Hock 2010 {published data only}
- Hock BD, McKenzie JL, McArthur L, Tansley S, Taylor KG, Fernyhough LJ. CD38 as a prognostic marker in chronic lymphocytic leukaemia at a single New Zealand centre: patient survival in comparison to age- and sex-matched population data. Internal Medicine Journal 2010;40(12):842-9. [DOI] [PubMed] [Google Scholar]
Houldsworth 2014 {published data only}
- Houldsworth J, Guttapalli A, Thodima V, Yan XJ, Mendiratta G, Zielonka T, et al. Genomic imbalance defines three prognostic groups for risk stratification of patients with chronic lymphocytic leukemia. Leukemia & Lymphoma 2014;55(4):920-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Hus 2006 {published data only}
- Hus I, Podhorecka M, Bojarska-Junak A, Rolinski J, Schmitt M, Sieklucka M, et al. The clinical significance of ZAP-70 and CD38 expression in B-cell chronic lymphocytic leukaemia. Annals of Oncology 2006;17(4):683-90. [DOI] [PubMed] [Google Scholar]
Jaksic 1981 {published data only}
- Jaksic B, Vitale B. Total tumour mass score (TTM): a new parameter in chronic lymphocyte leukaemia. British Journal of Haematology 1981;49(3):405-13. [DOI] [PubMed] [Google Scholar]
Jaksic 1992 {published data only}
- Jaksic B, Kusec R. More on early chronic lymphocytic leukemia (CLL): clinical staging systems and indications for treatment. Hematologic Pathology 1992;6(4):219-21. [PubMed] [Google Scholar]
Jaksic 2014 {published data only}
- Jaksic O, Vitale B, Jaksic B. An old and simple solution for a new problem - more on clinical staging and evaluation of response in B-cell chronic lymphocytic leukaemia in the era of new therapies. British Journal of Haematology 2014;165(5):737-40. [DOI] [PubMed] [Google Scholar]
Josefsson 2007 {published data only}
- Josefsson P, Geisler CH, Leffers H, Petersen JH, Andersen MK, Jurlander J, et al. CLLU1 expression analysis adds prognostic information to risk prediction in chronic lymphocytic leukemia. Blood 2007;109(11):4973-9. [DOI] [PubMed] [Google Scholar]
Juliusson 1986 {published data only}
- Juliusson G. Immunologic and cytogenetic studies improve prognosis prediction in chronic B-lymphocytic leukemia. A multivariate analysis of 24 variables. Cancer 1986;58(3):688-93. [DOI] [PubMed] [Google Scholar]
Juliusson 1990 {published data only}
- Juliusson G, Oscier DG, Fitchett M, Ross FM, Stockdill G, Mackie MJ, et al. Prognostic subgroups in B-cell chronic lymphocytic leukemia defined by specific chromosomal abnormalities. New England Journal of Medicine 1990;323(11):720-4. [DOI] [PubMed] [Google Scholar]
Kahraman 2014 {published data only}
- Kahraman Cetin N, Kacar Doger F, Kurtgoz S, Ruscuklu D, Yavasoglu I. Prognostic significance of the IgVH mutation status and immunohistochemical analysis of ZAP70 and CD38 in bone marrow biopsies in chronic lymphocytic leukemia. Turkiye Klinikleri Journal of Medical Sciences 2014;34(3):334-44. [Google Scholar]
Kardum‐Skelin 2008 {published data only}
- Kardum-Skelin I, Planinc-Peraica A, Ostojic Kolonic S, Radic-Kristo D, Milas M, Vrhovac R, et al. Clinical and laboratory prognostic parameters for leukemic types of chronic lymphoproliferative diseases. Acta Medica Croatica 2008;62(4):351-64. [PubMed] [Google Scholar]
Kardum‐Skelin 2009 {published data only}
- Kardum-Skelin I, Jaksic O, Kolonic SO, Vrhovac R, Fabijanic I, Jelic-Puskaric B, et al. New parameters of diploid histogram of image DNA cytometry and newly characterized types of nucleolar organizer region structures in defining the proliferative-kinetic index in chronic leukemic lymphoproliferative disorders. Analytical & Quantitative Cytology & Histology 2009;31(5):313-23. [PubMed] [Google Scholar]
Karmiris 1994 {published data only}
- Karmiris T, Rohatiner AZ, Love S, Carter M, Ganjoo RK, Amess J, et al. The management of chronic lymphocytic leukemia at a single centre over a 24-year period: prognostic factors for survival. Hematological Oncology 1994;12(1):29-39. [DOI] [PubMed] [Google Scholar]
Kay 2018 {published data only}
- Kay NE, LaPlant BR, Pettinger AM, Call TG, Leis JF, Ding W, et al. Cumulative experience and long term follow-up of pentostatin-based chemoimmunotherapy trials for patients with chronic lymphocytic leukemia. Expert Review of Haematology 2018;11(4):337-49. [DOI] [PubMed] [Google Scholar]
Keating 2000 {published data only}
- Keating MJ, Smith TL, Lerner S, O'Brien S, Robertson LE, Kantarjian H, et al. Prediction of prognosis following fludarabine used as secondary therapy for chronic lymphocytic leukemia. Leukemia and Lymphoma 2000;37(1-2):71-85. [DOI] [PubMed] [Google Scholar]
Khalifa 2002 {published data only}
- Khalifa M, Chehata S, Laatiri MA, Grira C, Gharbi O, Kortas M, et al. Epidemiologic, clinical and therapeutic aspects of chronic lymphoid leukemia: apropos of 120 cases. Tunisie Medicale 2002;80(10):584-9. [PubMed] [Google Scholar]
Kim 2004 {published data only}
- Kim SZ, Chow KU, Kukoc-Zivojnov N, Boehrer S, Brieger A, Steimle-Grauer SA, et al. Expression of ZAP-70 protein correlates with disease stage in chronic lymphocytic leukemia and is associated with, but not generally restricted to, non-mutated Ig VH status. Leukemia and Lymphoma 2004;45(10):2037-45. [DOI] [PubMed] [Google Scholar]
Kimby 1988 {published data only}
- Kimby E, Mellstedt H, Nilsson B, Bjorkholm M, Holm G, Lindemalm C, et al. Blood lymphocyte characteristics as predictors of prognosis in chronic lymphocytic leukemia of B-cell type. Hematological Oncology 1988;6(1):47-55. [DOI] [PubMed] [Google Scholar]
Kleinstern 2018 {published data only}
- Kleinstern G, Camp NJ, Goldin LR, Vachon CM, Vajdic CM, De Sanjose S, et al. Association of polygenic risk score with the risk of chronic lymphocytic leukemia and monoclonal B-cell lymphocytosis. Blood 2018;131:2541-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
Knospe 1977 {published data only}
- Knospe WH, Gregory SA, Trobaugh FE Jr, Stedronsky JA, Schrek R. Chronic lymphocytic leukemia: correlation of clinical course and therapeutic response with in vitro testing and morphology of lymphocytes. American Journal of Hematology 1977;2(1):73-101. [DOI] [PubMed] [Google Scholar]
Koberda 1989 {published data only}
- Koberda J, Czyz J, Hellmann A. Leukocyte doubling time as a prognostic factor in chronic lymphocytic leukemia. Acta Haematologica Polonica 1989;20(2):195-200. [PubMed] [Google Scholar]
Korycka‐Wolowiec 2011 {published data only}
- Korycka-Wolowiec A, Kotkowska A, Wawrzyniak E, Blonski JZ, Robak T. Do chromosomal aberrations detected in chronic lymphocytic leukemia patients by conventional cytogenetics with DSP30 have the clinical significance? Preliminary results. Acta Haematologica Polonica 2011;42(4):669-80. [Google Scholar]
Krober 2002 {published data only}
- Krober A, Seiler T, Benner A, Bullinger L, Bruckle E, Lichter P, et al. V(H) mutation status, CD38 expression level, genomic aberrations, and survival in chronic lymphocytic leukemia. Blood 2002;100(4):1410-6. [PubMed] [Google Scholar]
Krober 2006 {published data only}
- Krober A, Bloehdorn J, Hafner S, Buhler A, Seiler T, Kienle D, et al. Additional genetic high-risk features such as 11q deletion, 17p deletion, and V3-21 usage characterize discordance of ZAP-70 and VH mutation status in chronic lymphocytic leukemia. Journal of Clinical Oncology 2006;24(6):969-75. [DOI] [PubMed] [Google Scholar]
Kryachok 2011 {published data only}
- Kryachok I, Abramenko I, Bilous N, Chumak A, Martina Z, Filonenko I. IGHV gene rearrangements as outcome predictors for CLL patients: experience of Ukrainian group. Medical Oncology 2011;29(2):1093-101. [DOI] [PubMed] [Google Scholar]
Kurec 1992 {published data only}
- Kurec AS, Threatte GA, Gottlieb AJ, Smith JR, Anderson J, Davey FR. Immunophenotypic subclassification of chronic lymphocytic leukaemia (CLL). British Journal of Haematology 1992;81(1):45-51. [DOI] [PubMed] [Google Scholar]
Lai 2002 {published data only}
- Lai R, O'Brien S, Maushouri T, Rogers A, Kantarjian H, Keating M, et al. Prognostic value of plasma interleukin-6 levels in patients with chronic lymphocytic leukemia. Cancer 2002;95(5):1071-5. [DOI] [PubMed] [Google Scholar]
Lech‐Maranda 2012 {published data only}
- Lech-Maranda E, Grzybowska-Izydorczyk O, Wyka K, Mlynarski W, Borowiec M, Antosik K, et al. Serum tumor necrosis factor-alpha and interleukin-10 levels as markers to predict outcome of patients with chronic lymphocytic leukemia in different risk groups defined by the IGHV mutation status. Archivum Immunologiae et Therapiae Experimentalis 2012;60(6):477-86. [DOI] [PubMed] [Google Scholar]
Lech‐Maranda 2013 {published data only}
- Lech-Maranda E, Mlynarski W, Grzybowska-Izydorczyk O, Borowiec M, Pastorczak A, Cebula-Obrzut B, et al. Polymorphisms of TNF and IL-10 genes and clinical outcome of patients with chronic lymphocytic leukemia. Genes, Chromosomes & Cancer 2013;52(3):287-96. [DOI] [PubMed] [Google Scholar]
Lecouvet 1997 {published data only}
- Lecouvet FE, Vande Berg BC, Michaux L, Schmitz PJ, Malghem J, Jamart J, et al. Early chronic lymphocytic leukemia: prognostic value of quantitative bone marrow MR imaging findings and correlation with hematologic variables. Radiology 1997;204(3):813-8. [DOI] [PubMed] [Google Scholar]
Li 2008 {published data only}
- Li FJ, Ding S, Pan J, Shakhmatov MA, Kashentseva E, Wu J, et al. FCRL2 expression predicts IGHV mutation status and clinical progression in chronic lymphocytic leukemia. Blood 2008;112(1):179-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
Li 2017a {published data only}
- Li H, Xiong W, Liu H, Yi S, Yu Z, Liu W, et al. Serum LDH level may predict outcome of chronic lymphocytic leukemia patients with a 17p deletion: a retrospective analysis of prognostic factors in China. Chinese Journal of Cancer Research 2017;29(2):156-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
Lin 2002 {published data only}
- Lin K, Sherrington PD, Dennis M, Matrai Z, Cawley JC, Pettitt AR. Relationship between p53 dysfunction, CD38 expression, and IgV(H) mutation in chronic lymphocytic leukemia. Blood 2002;100(4):1404-9. [DOI] [PubMed] [Google Scholar]
Lin 2014 {published data only}
- Lin TT, Norris K, Heppel NH, Pratt G, Allan JM, Allsup DJ, et al. Telomere dysfunction accurately predicts clinical outcome in chronic lymphocytic leukaemia, even in patients with early stage disease. British Journal of Haematology 2014;167(2):214-23. [DOI] [PubMed] [Google Scholar]
Lozano‐Santos 2014 {published data only}
- Lozano-Santos C, Martinez-Velasquez J, Fernandez-Cuevas B, Polo N, Navarro B, Millan I, et al. Vascular endothelial growth factor A (VEGFA) gene polymorphisms have an impact on survival in a subgroup of indolent patients with chronic lymphocytic leukemia. PLOS One 2014;9(6):e101063. [DOI] [PMC free article] [PubMed] [Google Scholar]
Lucas 2015 {published data only}
- Lucas DM, Ruppert AS, Lozanski G, Dewald GW, Lozanski A, Claus R, et al. Cytogenetic prioritization with inclusion of molecular markers predicts outcome in previously untreated patients with chronic lymphocytic leukemia treated with fludarabine or fludarabine plus cyclophosphamide: a long-term follow-up study of the US intergroup phase III trial E2997. Leukemia and Lymphoma 2015;56(11):3031-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
Maffei 2007 {published data only}
- Maffei R, Marasca R, Martinelli S, Castelli I, Santachiara R, Morandi E, et al. Angiopoietin-2 expression in B-cell chronic lymphocytic leukemia: association with clinical outcome and immunoglobulin heavy-chain mutational status. Leukemia 2007;21(6):1312-5. [DOI] [PubMed] [Google Scholar]
Maffei 2010 {published data only}
- Maffei R, Martinelli S, Santachiara R, Rossi D, Guarnotta C, Sozzi E, et al. Angiopoietin-2 plasma dosage predicts time to first treatment and overall survival in chronic lymphocytic leukemia. Blood 2010;116(4):584-92. [DOI] [PubMed] [Google Scholar]
Mandelli 1987 {published data only}
- Mandelli F, De Rossi G, Mancini P, Alberti A, Cajozzo A, Grignani F, et al. Prognosis in chronic lymphocytic leukemia: a retrospective multicentric study from the GIMEMA group. Journal of Clinical Oncology 1987;5(3):398-406. [DOI] [PubMed] [Google Scholar]
Mansouri 2013 {published data only}
- Mansouri L, Grabowski P, Degerman S, Svenson U, Gunnarsson R, Cahill N, et al. Short telomere length is associated with NOTCH1/SF3B1/TP53 aberrations and poor outcome in newly diagnosed chronic lymphocytic leukemia patients. American Journal of Hematology 2013;88(8):647-51. [DOI] [PubMed] [Google Scholar]
Marasca 2005 {published data only}
- Marasca R, Maffei R, Morselli M, Zucchini P, Castelli I, Martinelli S, et al. Immunoglobulin mutational status detected through single-round amplification of partial V(H) region represents a good prognostic marker for clinical outcome in chronic lymphocytic leukemia. Journal of Molecular Diagnostics 2005;7(5):566-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
Marasca 2013 {published data only}
- Marasca R, Maffei R, Martinelli S, Fiorcari S, Bulgarelli J, Debbia G, et al. Clinical heterogeneity of de novo 11q deletion chronic lymphocytic leukaemia: prognostic relevance of extent of 11q deleted nuclei inside leukemic clone. Hematological Oncology 2013;31(2):348-55. [DOI] [PubMed] [Google Scholar]
Martinelli 2008 {published data only}
- Martinelli S, Maffei R, Castelli I, Santachiara R, Zucchini P, Fontana M, et al. Increased expression of angiopoietin-2 characterizes early B-cell chronic lymphocytic leukemia with poor prognosis. Leukemia Research 2008;32(4):593-7. [DOI] [PubMed] [Google Scholar]
Masic 1998 {published data only}
- Masic N, Gagro A, Rabatic S, Sabioncello A, Dasic G, Jaksic B, et al. Decision-tree approach to the immunophenotype-based prognosis of the B-cell chronic lymphocytic leukemia. American Journal of Hematology 1998;59(2):143-8. [DOI] [PubMed] [Google Scholar]
Mateva 2001 {published data only}
- Mateva N, Nenova I, Dimitrov I, Karnolski I, Ananoshtev N. A predictive model for survival of patients with chronic lymphocytic leukemia - results of a multivariate survival analysis. Journal of the Balkan Union of Oncology 2001;6(4):429-33. [Google Scholar]
Matthews 2006 {published data only}
- Matthews C, Catherwood MA, Morris TC, Kettle PJ, Drake MB, Gilmore WS, et al. Serum TK levels in CLL identify Binet stage A patients within biologically defined prognostic subgroups most likely to undergo disease progression. European Journal of Haematology 2006;77(4):309-17. [DOI] [PubMed] [Google Scholar]
Matthews 2007 {published data only}
- Matthews C, Catherwood MA, Morris TC, Alexander HD. V(H)3-48 and V(H)3-53, as well as V(H)3-21, gene rearrangements define unique subgroups in CLL and are associated with biased lambda light chain restriction, homologous LCDR3 sequences and poor prognosis. Leukemia Research 2007;31(2):231-4. [DOI] [PubMed] [Google Scholar]
Matutes 2013 {published data only}
- Matutes E, Bosanquet AG, Wade R, Richards SM, Else M, Catovsky D. The use of individualized tumor response testing in treatment selection: second randomization results from the LRF CLL4 trial and the predictive value of the test at trial entry. Leukemia 2013;27(2):507-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
Matutes 2017 {published data only}
- Matutes E, Polliack A. Predicting time to first treatment in early stage CLL: scores, values and comparative prognostic models. Leukemia & Lymphoma 2017;58(7):1528-9. [DOI] [PubMed] [Google Scholar]
Melo 1987 {published data only}
- Melo JV, Catovsky D, Gregory WM, Galton DA. The relationship between chronic lymphocytic leukaemia and prolymphocytic leukaemia. IV. Analysis of survival and prognostic features. British Journal of Haematology 1987;65(1):23-9. [DOI] [PubMed] [Google Scholar]
Miao 2018 {published data only}
- Miao Y, Miao Y, Shi K, Sun Q, Zhao SS, Xia Y, et al. A higher percentage of cells with 13q deletion predicts worse outcome in Chinese patients with chronic lymphocytic leukemia carrying isolated 13q deletion. Annals of Hematology 2018;97:1663–9. [DOI] [PubMed] [Google Scholar]
Molica 1984 {published data only}
- Molica S, Puzzonia P, Iannaccaro P, Pane C, Alberti A. Prognosis of chronic lymphocytic leukemia: analysis of one hundred cases. Tumori 1984;70(5):399-402. [DOI] [PubMed] [Google Scholar]
Molica 1986 {published data only}
- Molica S, Alberti A. Prognostic value of "total tumor mass score" (TTM): a retrospective analysis of 130 patients with chronic lymphocytic leukemia. Tumori 1986;72(6):559-64. [DOI] [PubMed] [Google Scholar]
Molica 1988 {published data only}
- Molica S, Alberti A. Investigation of nuclear clefts as a prognostic parameter in chronic lymphocytic leukemia. European Journal of Haematology 1988;41(1):62-5. [DOI] [PubMed] [Google Scholar]
Molica 1991 {published data only}
- Molica S. Progression and survival studies in early chronic lymphocytic leukemia. Blood 1991;78(4):895-9. [PubMed] [Google Scholar]
Molica 1994 {published data only}
- Molica S, Brugiatelli M, Callea V, Morabito F, Levato D, Nobile F, et al. Comparison of younger versus older B-cell chronic lymphocytic leukemia patients for clinical presentation and prognosis. A retrospective study of 53 cases. European Journal of Haematology 1994;52(4):216-21. [DOI] [PubMed] [Google Scholar]
Molica 1998 {published data only}
- Molica S, Levato D, Dattilo A, Mannella A. Clinico-prognostic relevance of quantitative immunophenotyping in B-cell chronic lymphocytic leukemia with emphasis on the expression of CD20 antigen and surface immunoglobulins. European Journal of Haematology 1998;60(1):47-52. [DOI] [PubMed] [Google Scholar]
Molica 1999a {published data only}
- Molica S, Levato D, Cascavilla N, Levato L, Musto P. Clinico-prognostic implications of simultaneous increased serum levels of soluble CD23 and beta2-microglobulin in B-cell chronic lymphocytic leukemia. European Journal of Haematology 1999;62(2):117-22. [DOI] [PubMed] [Google Scholar]
Molica 1999b {published data only}
- Molica S, Levato D, Dattilo A. Natural history of early chronic lymphocytic leukemia. A single institution study with emphasis on the impact of disease-progression on overall survival. Haematologica 1999;84(12):1094-9. [PubMed] [Google Scholar]
Molica 2008 {published data only}
- Molica S, Vitelli G, Cutrona G, Todoerti K, Mirabelli R, Digiesi G, et al. Serum thrombopoietin compared with ZAP-70 and immunoglobulin heavy-chain gene mutation status as a predictor of time to first treatment in early chronic lymphocytic leukemia. Leukemia and Lymphoma 2008;49(1):62-7. [DOI] [PubMed] [Google Scholar]
Montserrat 1991 {published data only}
- Montserrat E, Gomis F, Vallespi T, Rios A, Romero A, Soler J, et al. Presenting features and prognosis of chronic lymphocytic leukemia in younger adults. Blood 1991;78(6):1545-51. [PubMed] [Google Scholar]
Morabito 2001 {published data only}
- Morabito F, Mangiola M, Oliva B, Stelitano C, Callea V, Deaglio S, et al. Peripheral blood CD38 expression predicts survival in B-cell chronic lymphocytic leukemia. Leukemia Research 2001;25(11):927-32. [DOI] [PubMed] [Google Scholar]
Morabito 2015a {published data only}
- Morabito F, Cutrona G, Gentile M, Fabris S, Matis S, Vigna E, et al. Is ZAP70 still a key prognostic factor in early stage chronic lymphocytic leukaemia? Results of the analysis from a prospective multicentre observational study. British Journal of Haematology 2015;168(3):455-9. [DOI] [PubMed] [Google Scholar]
Morabito 2015b {published data only}
- Morabito F, Cutrona G, Mosca L, D'Anca M, Matis S, Gentile M, et al. Surrogate molecular markers for IGHV mutational status in chronic lymphocytic leukemia for predicting time to first treatment. Leukemia Research 2015;39(8):840-5. [DOI] [PubMed] [Google Scholar]
Morabito 2018c {published data only}
- Morabito F, Shanafelt TD, Gentile M, Reda G, Mauro FR, Rossi D, et al. Immunoglobulin heavy chain variable region gene and prediction of time to first treatment in patients with chronic lymphocytic leukemia: mutational load or mutational status? Analysis of 1003 cases. American Journal of Hematology 2018;93(9):E216-9. [DOI] [PubMed] [Google Scholar]
Moreno 2019 {published data only}
- Moreno C, Delgado J, Byrd JC, Zvagelsky WL, Suzuki S, Hsu E, et al. Changes in clinical stage identify patients with CLL and different outcome within iwCLL partial response: RESONATE study. British Journal of Haematology 2019;185(1):148-50. [DOI: 10.1111/bjh.15397] [DOI] [PMC free article] [PubMed] [Google Scholar]
Morilla 2008 {published data only}
- Morilla A, Gonzalez de Castro D, Del Giudice I, Osuji N, Else M, Morilla R, et al. Combinations of ZAP-70, CD38 and IGHV mutational status as predictors of time to first treatment in CLL. Leukemia & Lymphoma 2008;49(11):2108-15. [DOI] [PubMed] [Google Scholar]
Nabhan 2017 {published data only}
- Nabhan C, Mato A, Flowers CR, Grinblatt DL, Lamanna N, Weiss MA, et al. Characterizing and prognosticating chronic lymphocytic leukemia in the elderly: prospective evaluation on 455 patients treated in the United States. BMC Cancer 2017;17(1):198. [DOI] [PMC free article] [PubMed] [Google Scholar]
Nedeva 2018 {published data only}
- Nedeva A, Naseva E, Kindekov I, Petkova N, Nikolov I, Raynov J. The prognostic significance of interphase cytogenetic abnormalities in chronic lymphocytic leukemia. Archives of Hellenic Medicine 2018;35(4):520-6. [Google Scholar]
Nenova 2000 {published data only}
- Nenova I, Karnolski I, Mateva N, Ananoshtev N. A study of prognostic factors in patients with chronic lymphocytic leukemia. Journal of Bulkan Union of Oncology 2000;5(1):49-54. [Google Scholar]
Nipp 2014 {published data only}
- Nipp RD, Volkheimer AD, Davis ED, Chen Y, Weinberg JB, Friedman DR. CD38 variation as a prognostic factor in chronic lymphocytic leukemia. Leukemia & Lymphoma 2014;55(1):191-4. [DOI] [PubMed] [Google Scholar]
Nola 2004 {published data only}
- Nola M, Pavletic SZ, Weisenburger DD, Smith LM, Bast MA, Vose JM, et al. Prognostic factors influencing survival in patients with B-cell small lymphocytic lymphoma. American Journal of Hematology 2004;77(1):31-5. [DOI] [PubMed] [Google Scholar]
Nowakowski 2009 {published data only}
- Nowakowski GS, Hoyer JD, Shanafelt TD, Zent CS, Call TG, Bone ND, et al. Percentage of smudge cells on routine blood smear predicts survival in chronic lymphocytic leukemia. Journal of Clinical Oncology 2009;27(11):1844-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Nuckel 2006 {published data only}
- Nuckel H, Huttmann A, Klein-Hitpass L, Schroers R, Fuhrer A, Sellmann L, et al. Lipoprotein lipase expression is a novel prognostic factor in B-cell chronic lymphocytic leukemia. Leukemia & Lymphoma 2006;47(6):1053-61. [DOI] [PubMed] [Google Scholar]
Nuckel 2009 {published data only}
- Nuckel H, Collins CH, Frey UH, Sellmann L, Durig J, Siffert W, et al. FCRL2 mRNA expression is inversely associated with clinical progression in chronic lymphocytic leukemia. European Journal of Haematology 2009;83(6):541-9. [DOI] [PubMed] [Google Scholar]
O'Brien 1993 {published data only}
- O'Brien S, Kantarjian H, Beran M, Smith T, Koller C, Estey E, et al. Results of fludarabine and prednisone therapy in 264 patients with chronic lymphocytic leukemia with multivariate analysis-derived prognostic model for response to treatment. Blood 1993;82(6):1695-700. [PubMed] [Google Scholar]
Ocana 2007 {published data only}
- Ocana E, Delgado-Perez L, Campos-Caro A, Munoz J, Paz A, Franco R, et al. The prognostic role of CXCR3 expression by chronic lymphocytic leukemia B cells. Haematologica 2007;92(3):349-56. [DOI] [PubMed] [Google Scholar]
Oliveira 2011 {published data only}
- Oliveira AC, De La Banda E, Domingo-Domenech E, Encuentra M, Mercadal S, Domingo A, et al. Prospective study of clinical and biological prognostic factors at diagnosis in patients with early stage B-cell chronic lymphocytic leukemia. Leukemia and Lymphoma 2011;52(3):429-35. [DOI] [PubMed] [Google Scholar]
Orgueira 2019 {published data only}
- Orgueira AM, Rodriguez BA, Vence NA, Lopez AB, Arias JA, Varela ND, et al. Time to treatment prediction in chronic lymphocytic leukemia based on new transcriptional patterns. Frontiers in Oncology 2019;9:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
Oscier 1990 {published data only}
- Oscier DG, Stevens J, Hamblin TJ, Pickering RM, Lambert R, Fitchett M. Correlation of chromosome abnormalities with laboratory features and clinical course in B-cell chronic lymphocytic leukaemia. British Journal of Haematology 1990;76(3):352-8. [DOI] [PubMed] [Google Scholar]
Paolino 1984 {published data only}
- Paolino W, Infelise V, Levis A, Marmont F, Vitolo U, Paolino F, et al. Adenosplenomegaly and prognosis in uncomplicated and complicated chronic lymphocytic leukemia. A study of 362 cases. Cancer 1984;54(2):339-46. [DOI] [PubMed] [Google Scholar]
Plesingerova 2017 {published data only}
- Plesingerova H, Librova Z, Plevova K, Libra A, Tichy B, Skuhrova Francova H, et al. COBLL1, LPL and ZAP70 expression defines prognostic subgroups of chronic lymphocytic leukemia patients with high accuracy and correlates with IGHV mutational status. Leukemia & Lymphoma 2017;58(1):70-9. [DOI] [PubMed] [Google Scholar]
Prokocimer 1985 {published data only}
- Prokocimer M, Modan M, Lusky A, Hershko C. Multivariate analysis of prognostic factors in chronic lymphocytic leukemia. Israel Journal of Medical Sciences 1985;21(6):490-8. [PubMed] [Google Scholar]
Qin 2017 {published data only}
- Qin SC, Xia Y, Miao Y, Zhu HY, Wu JZ, Fan L, et al. MYD88 mutations predict unfavorable prognosis in chronic lymphocytic leukemia patients with mutated IGHV gene. Blood Cancer Journal 2017;7(12):651. [DOI] [PMC free article] [PubMed] [Google Scholar]
Queiros 2015 {published data only}
- Queiros AC, Villamor N, Clot G, Martinez-Trillos A, Kulis M, Navarro A, et al. A B-cell epigenetic signature defines three biologic subgroups of chronic lymphocytic leukemia with clinical impact. Leukemia 2015;29(3):598-605. [DOI] [PubMed] [Google Scholar]
Rai 1975 {published data only}
- Rai KR, Sawitsky A, Cronkite EP, Chanana AD, Levy RN, Pasternack BS. Clinical staging of chronic lymphocytic leukemia. Blood 1975;46(2):219-34. [DOI] [PubMed] [Google Scholar]
Rai 1990 {published data only}
- Rai KR, Han T. Prognostic factors and clinical staging in chronic lymphocytic leukemia. Hematology/Oncology Clinics of North America 1990;4(2):447-56. [PubMed] [Google Scholar]
Raponi 2018 {published data only}
- Raponi S, Del Giudice I, Marinelli M, Wang J, Cafforio L, Ilari C, et al. Genetic landscape of ultra-stable chronic lymphocytic leukemia patients. Annals of Oncology 2018;29(4):966-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
Resegotti 1989 {published data only}
- Resegotti L, Levis A, Decrescenzo A, Ficara F, Pini M. Significance and independence of the immunologic, histological, and clinical features as prognostic factors in chronic lymphatic leukemia. Haematologica 1989;74(5 Suppl):368-79. [PubMed] [Google Scholar]
Rissiek 2014 {published data only}
- Rissiek A, Schulze C, Bacher U, Schieferdecker A, Thiele B, Jacholkowski A, et al. Multidimensional scaling analysis identifies pathological and prognostically relevant profiles of circulating T-cells in chronic lymphocytic leukemia. International Journal of Cancer 2014;135(10):2370-9. [DOI] [PubMed] [Google Scholar]
Rodriguez 2007 {published data only}
- Rodriguez A, Villuendas R, Yanez L, Gomez ME, Diaz R, Pollan M, et al. Molecular heterogeneity in chronic lymphocytic leukemia is dependent on BCR signaling: clinical correlation. Leukemia 2007;21(9):1984-91. [DOI] [PubMed] [Google Scholar]
Ronchetti 2016 {published data only}
- Ronchetti D, Manzoni M, Agnelli L, Vinci C, Fabris S, Cutrona G, et al. lncRNA profiling in early-stage chronic lymphocytic leukemia identifies transcriptional fingerprints with relevance in clinical outcome. Blood Cancer Journal 2016;6(9):e468. [DOI] [PMC free article] [PubMed] [Google Scholar]
Rossi 1986 {published data only}
- Rossi PL, Alfano G, D'Onofrio G, Menichella G, Mango G. Depletion lymphocytapheresis in chronic lymphocytic leukemias: criteria for predicting which patients will respond to treatment. International Journal of Artificial Organs 1986;9(1):59-62. [PubMed] [Google Scholar]
Rossi 2010a {published data only}
- Rossi FM, Del Principe MI, Rossi D, Irno Consalvo M, Luciano F, Zucchetto A, et al. Prognostic impact of ZAP-70 expression in chronic lymphocytic leukemia: mean fluorescence intensity T/B ratio versus percentage of positive cells. Journal of Translational Medicine 2010;8:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
Rossi 2010b {published data only}
- Rossi S, Shimizu M, Barbarotto E, Nicoloso MS, Dimitri F, Sampath D, et al. MicroRNA fingerprinting of CLL patients with chromosome 17p deletion identify a miR-21 score that stratifies early survival. Blood 2010;116(6):945-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
Rossi 2011 {published data only}
- Rossi D, Spina V, Deambrogi C, Rasi S, Laurenti L, Stamatopoulos K, et al. The genetics of Richter syndrome reveals disease heterogeneity and predicts survival after transformation. Blood 2011;117(12):3391-401. [DOI] [PubMed] [Google Scholar]
Rozman 1979 {published data only}
- Rozman C, Montserrat Costa E, Morey M, Aranalde JM, Feliu E, Granena A, et al. Chronic lymphoid leukemia. Survival in relation to clinical stages. Statistical analysis of 95 cases. Medicina Clinica 1979;72(7):265-71. [PubMed] [Google Scholar]
Salomon‐Nguyen 1995 {published data only}
- Salomon-Nguyen F, Valensi F, Merle-Beral H, Flandrin G. A scoring system for the classification of CD5-B CLL versus CD5+ B CLL and B PLL. Leukemia & Lymphoma 1995;16(5-6):445-50. [DOI] [PubMed] [Google Scholar]
Santoro 1979 {published data only}
- Santoro A, Musumeci R, Rilke F, Franchi F, Valagussa P, Bajetta E, et al. Clinical classification and survival in chronic lymphocyte leukemia. Tumori 1979;65(1):39-49. [DOI] [PubMed] [Google Scholar]
Sarmiento 2002 {published data only}
- Sarmiento MA, Palacios MF, Scolnik MP, Ramirez FR, Stanganelli C, Cabrera J, et al. Evolution of chronic lymphocytic leukemia: predictive value of immunophenotype, soluble CD23 and morphology. Medicina 2002;62(4):305-12. [PubMed] [Google Scholar]
Savvopoulos 2016 {published data only}
- Savvopoulos S, Misener R, Panoskaltsis N, Pistikopoulos EN, Mantalaris A. A personalized framework for dynamic modeling of disease trajectories in chronic lymphocytic leukemia. IEEE Transactions on Biomedical Engineering 2016;63(11):2396-404. [DOI] [PubMed] [Google Scholar]
Scolozzi 1981 {published data only}
- Scolozzi R, Boccafogli A, Romanini D, Ramazzina E, Careri C, Guidoboni CA. Prognosis of chronic lymphatic leukemia. Retrospective study of the survival period 47 patients. Minerva Medica 1981;72(4-5):205-11. [PubMed] [Google Scholar]
Shanafelt 2010 {published data only}
- Shanafelt TD, Rabe KG, Kay NE, Zent CS, Jelinek DF, Reinalda MS, et al. Age at diagnosis and the utility of prognostic testing in patients with chronic lymphocytic leukemia. Cancer 2010;116(20):4777-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
Shanafelt 2017 {published data only}
- Shanafelt TD, Parikh SA, Noseworthy PA, Goede V, Chaffee KG, Bahlo J, et al. Atrial fibrillation in patients with chronic lymphocytic leukemia (CLL). Leukemia & Lymphoma 2017;58(7):1630-9. [DOI] [PubMed] [Google Scholar]
Spacek 2009 {published data only}
- Spacek M, Pekova S, Bezdickova L, Kozak T. Risk factors in chronic lymphocytic leukemia, validation of flow cytometric detection using RT-PCR and immunohistochemistry. Transfuze a Hematologie Dnes 2009;15(2):91-6. [Google Scholar]
Stamatopoulos 2009 {published data only}
- Stamatopoulos B, Meuleman N, Haibe-Kains B, Saussoy P, Van Den Neste E, Michaux L, et al. MicroRNA-29c and microRNA-223 down-regulation has in vivo significance in chronic lymphocytic leukemia and improves disease risk stratification. Blood 2009;113(21):5237-45. [DOI] [PubMed] [Google Scholar]
Stamatopoulos 2017 {published data only}
- Stamatopoulos B, Timbs A, Bruce D, Smith T, Clifford R, Robbe P, et al. Targeted deep sequencing reveals clinically relevant subclonal IgHV rearrangements in chronic lymphocytic leukemia. Leukemia 2017;31(4):837-45. [DOI] [PubMed] [Google Scholar]
Strefford 2015 {published data only}
- Strefford JC, Kadalayil L, Forster J, Rose-Zerilli MJ, Parker A, Lin TT, et al. Telomere length predicts progression and overall survival in chronic lymphocytic leukemia: data from the UK LRF CLL4 trial. Leukemia 2015;10(10):217. [DOI] [PMC free article] [PubMed] [Google Scholar]
Szymczyk 2018 {published data only}
- Szymczyk A, Chocholska S, Macheta A, Szczepanek D, Hus M, Podhorecka M. Assessment of micro RNAs expression in leukemic cells as prognostic markers in chronic lymphocytic leukemia: micro RNAs can predict survival in a course of the disease. Oncotarget 2018;9(27):19136-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
Tallarico 2018 {published data only}
- Tallarico M, Foster JC, Seisler D, Lafky JM, Hurria A, Jatoi A, et al. Frequency and impact of grade three or four toxicities of novel agents on outcomes of older patients with chronic lymphocytic leukemia and non-Hodgkin lymphoma (alliance A151611). Journal of Geriatric Oncology 2018;9:321-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Tobin 2005a {published data only}
- Tobin G. The immunoglobulin genes and chronic lymphocytic leukemia (CLL). Upsala Journal of Medical Sciences 2005;110(2):97-113. [DOI] [PubMed] [Google Scholar]
Tobin 2005b {published data only}
- Tobin G, Thunberg U, Laurell A, Karlsson K, Aleskog A, Willander K, et al. Patients with chronic lymphocytic leukemia with mutated VH genes presenting with Binet stage B or C form a subgroup with a poor outcome. Haematologica 2005;90(4):465-9. [PubMed] [Google Scholar]
Vallat 2013 {published data only}
- Vallat L, Kemper CA, Jung N, Maumy-Bertrand M, Bertrand F, Meyer N, et al. Reverse-engineering the genetic circuitry of a cancer cell with predicted intervention in chronic lymphocytic leukemia. Proceedings of the National Academy of Sciences of the United States of America 2013;110(2):459-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
Van Damme 2012 {published data only}
- Van Damme M, Crompot E, Meuleman N, Mineur P, Bron D, Lagneaux L, et al. HDAC isoenzyme expression is deregulated in chronic lymphocytic leukemia B-cells and has a complex prognostic significance. Epigenetics 2012;7(12):1403-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
Velardi 1980 {published data only}
- Velardi A, Spinozzi F, Siracusa A, Colozza M, Aversa F, Rambotti P, et al. Clinical staging of chronic lymphocytic leukaemia and its relationship to survival. Haematologica 1980;65(1):82-95. [PubMed] [Google Scholar]
Vojdeman 2017 {published data only}
- Vojdeman FJ, Herman SE, Kirkby N, Wiestner A, Van T' Veer MB, Tjonnfjord GE, et al. Soluble CD52 is an indicator of disease activity in chronic lymphocytic leukemia. Leukemia & Lymphoma 2017;58(10):2356-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
Vural 2014 {published data only}
- Vural F, Karaca E, Soyer N, Gunduz C, Sahin F, Kosova B, et al. Comparison of CD38, ZAP70 and hTERT expression with known prognostic markers in patients with chronic lymphocytic leukemia during five-year follow- up period. Uluslararasi Hematoloji-Onkoloji Dergisi (UHOD) 2014;24(3):179-84. [Google Scholar]
Weinberg 2007 {published data only}
- Weinberg JB, Volkheimer AD, Chen Y, Beasley BE, Jiang N, Lanasa MC, et al. Clinical and molecular predictors of disease severity and survival in chronic lymphocytic leukemia. American Journal of Hematology 2007;82(12):1063-70 [Erratum appears in American Journal of Hematology 2008;83(1):90]. [DOI] [PubMed] [Google Scholar]
Weiss 2011 {published data only}
- Weiss L, Melchardt T, Egle A, Grabmer C, Greil R, Tinhofer I. Regulatory T cells predict the time to initial treatment in early stage chronic lymphocytic leukemia. Cancer 2011;117(10):2163-9. [DOI] [PubMed] [Google Scholar]
Wierda 2003 {published data only}
- Wierda WG, Johnson MM, Do KA, Manshouri T, Dey A, O'Brien S, et al. Plasma interleukin 8 level predicts for survival in chronic lymphocytic leukaemia. British Journal of Haematology 2003;120(3):452-6. [DOI] [PubMed] [Google Scholar]
Winkler 2010 {published data only}
- Winkler D, Schneider C, Zucknick M, Bogelein D, Schulze K, Zenz T, et al. Protein expression analysis of chronic lymphocytic leukemia defines the effect of genetic aberrations and uncovers a correlation of CDK4, P27 and P53 with hierarchical risk. Haematologica 2010;95(11):1880-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Wu 2010 {published data only}
- Wu T, Li ZJ, Wang YF, Qiu LG. Applicability of different clinical staging systems in chronic lymphocytic leukemia for Chinese patients. Journal of Leukemia and Lymphoma 2010;19(3):136-9. [Google Scholar]
Zengin 1997 {published data only}
- Zengin N, Kars A, Kansu E, Ozdemir O, Barista I, Gullu I, et al. Comparison of Rai and Binet classifications in chronic lymphocytic leukemia. Hematology 1997;2(2):125-9. [DOI] [PubMed] [Google Scholar]
Zenz 2009 {published data only}
- Zenz T, Benner A, Duhrsen U, Durig J, Dohner H, Siffert W, et al. BCL2-938C>A polymorphism and disease progression in chronic lymphocytic leukemia. Leukemia & Lymphoma 2009;50(11):1837-42. [DOI] [PubMed] [Google Scholar]
Zucchetto 2006 {published data only}
- Zucchetto A, Bomben R, Dal Bo M, Sonego P, Nanni P, Rupolo M, et al. A scoring system based on the expression of six surface molecules allows the identification of three prognostic risk groups in B-cell chronic lymphocytic leukemia. Journal of Cellular Physiology 2006;207(2):354-63. [DOI] [PubMed] [Google Scholar]
References to studies awaiting assessment
Antic 2011 {published data only}
- Antic D, Mihaljevic B, Cokic V, Fekete MD, Djurasevic TK, Pavlovic S, et al. Patients with early stage chronic lymphocytic leukemia: new risk stratification based on molecular profiling. Leukemia & Lymphoma 2011;52(7):1394-7. [DOI] [PubMed] [Google Scholar]
Baumann 2014 {published data only}
- Baumann T, Delgado J, Santacruz R, Martínez-Trillos A, Royo C, Navarro A, et al. Chronic lymphocytic leukemia in the elderly: clinico-biological features, outcomes, and proposal of a prognostic model. Haematologica 2014;99(10):1599-604. [DOI] [PMC free article] [PubMed] [Google Scholar]
Bulian 2011 {published data only}
- Bulian P, Tarnani M, Rossi D, Forconi F, Del Poeta G, Bertoni F, et al. Multicentre validation of a prognostic index for overall survival in chronic lymphocytic leukaemia. Hematological Oncology 2011;29(2):91-9. [DOI] [PubMed] [Google Scholar]
Bulian 2012 {published data only}
- Bulian P, Rossi D, Forconi F, Del Poeta G, Bertoni F, Zucca E, et al. IGHV gene mutational status and 17p deletion are independent molecular predictors in a comprehensive clinical-biological prognostic model for overall survival prediction in chronic lymphocytic leukemia. Journal of Translational Medicine 2012;10:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
Cavallini 2017 {published data only}
- Cavallini C, Visco C, Putta S, Rossi D, Mimiola E, Purvis N, et al. Integration of B-cell receptor-induced ERK1/2 phosphorylation and mutations of SF3B1 gene refines prognosis in treatment-naïve chronic lymphocytic leukemia. Haematologica 2017;102(4):e144-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
Del Guidice 2005 {published data only}
- Del Giudice I, Morilla A, Osuji N, Matutes E, Morilla R, Burford A, et al. Zeta-chain associated protein 70 and CD38 combined predict the time to first treatment in patients with chronic lymphocytic leukemia. Cancer 2005;104(10):2124-32. [DOI] [PubMed] [Google Scholar]
Friedrichs 2011 {published data only}
- Friedrichs B, Siegel S, Reimer R, Barsoum A, Coggin J Jr, Kabelitz D, et al. High expression of the immature laminin receptor protein correlates with mutated IGVH status and predicts a favorable prognosis in chronic lymphocytic leukemia. Leukemia Research 2011;35(6):721-9. [DOI] [PubMed] [Google Scholar]
Furundarena 1994 {published data only}
- Furundarena Salsamendi JR, Martinez D, Navarro E, Aguinaco R, Vidal MJ, Lasa R, et al. Chronic lymphocytic leukemia: a retrospective analysis of diagnostic characteristics and survival predictive factors in 150 patients. Biologia y Clinica Hematologica 1994;16(2):64-70. [Google Scholar]
Gentile 2009 {published data only}
- Gentile M, Cutrona G, Neri A, Molica S, Ferrarini M, Morabito F. Predictive value of beta2-microglobulin (beta2-m) levels in chronic lymphocytic leukemia since Binet A stages. Haematologica 2009;94(6):887-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
Haferlach 2010 {published data only}
- Haferlach C, Dicker F, Weiss T, Schnittger S, Beck C, Grote-Metke A, et al. Toward a comprehensive prognostic scoring system in chronic lymphocytic leukemia based on a combination of genetic parameters. Genes, Chromosomes & Cancer 2010;49(9):851-9. [DOI] [PubMed] [Google Scholar]
Jarque 1991 {published data only}
- Jarque I, Sanz G, Gomis F, Martinez J, Martin G, De la Rubia J, et al. Chronic lymphatic leukemia. II. Analysis of prognostic factors and development of survival predicting models. Study of 187 patients. Sangre 1991;36(4):285-94. [PubMed] [Google Scholar]
Lee 1987 {published data only}
- Lee JS, Dixon DO, Kantarjian HM, Keating MJ, Talpaz M. Prognosis of chronic lymphocytic leukemia: a multivariate regression analysis of 325 untreated patients. Blood 1987;69(3):929-36. [PubMed] [Google Scholar]
Leotard 2000 {published data only}
- Leotard S, Chastang C, Travade P, Jaudon MC, Tournilhac O, Baudet S, et al. Prognostic relevance of a scoring system based on clinical and biological parameters in early chronic lymphocytic leukemia. Hematology Journal 2000;1(5):301-6. [DOI] [PubMed] [Google Scholar]
Letestu 2010 {published data only}
- Letestu R, Levy V, Eclache V, Baran-Marszak F, Vaur D, Naguib D, et al. Prognosis of Binet stage A chronic lymphocytic leukemia patients: the strength of routine parameters. Blood 2010;116(22):4588-90. [DOI] [PubMed] [Google Scholar]
Li 2017b {published data only}
- Li H, Yi SH, Xiong WJ, Liu HM, Lyu R, Wang TY, et al. Chronic lymphocytic leukemia prognostic index: a new integrated scoring system to predict the Time to First Treatment in Chinese patients with chronic lymphocytic leukemia. Chinese Medical Journal 2017;130(2):135-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
Liang 2018 {published data only}
- Liang JH, Gao R, Dai JC, Gale RP, Li W, Fan L, et al. The prognostic role of HBV infection in chronic lymphocytic leukemia. Journal of Cancer Research and Clinical Oncology 2018;144:1309–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
Metze 2000 {published data only}
- Metze K, Lobo AM, Lorand-Metze I. Nucleolus organizer regions (AgNORs) and total tumor mass are independent prognostic parameters for treatment-free period in chronic lymphocytic leukemia. International Journal of Cancer 2000;89(5):440-3. [DOI] [PubMed] [Google Scholar]
Miao 2019 {published data only}
- Miao Y, Zou YX, Gu DL, Zhu HC, Zhu HY, Wang L, et al. SF3B1 mutation predicts unfavorable treatment-free survival in Chinese chronic lymphocytic leukemia patients. Annals of Translational Medicine 2019;7(8):176. [DOI] [PMC free article] [PubMed] [Google Scholar]
Molica 1990 {published data only}
- Molica S, Reverter JC, Alberti A, Montserrat E. Timing of diagnosis and lymphocyte accumulation patterns in chronic lymphocytic leukemia: analysis of their clinical significance. European Journal of Haematology 1990;44(5):277-81. [DOI] [PubMed] [Google Scholar]
Molica 2010 {published data only}
- Molica S, Digiesi G, Battaglia C, Cutrona G, Antenucci A, Molica M, et al. Baff serum level predicts time to first treatment in early chronic lymphocytic leukemia. European Journal of Haematology 2010;85(4):314-20. [DOI] [PubMed] [Google Scholar]
Molica 2015 {published data only}
- Molica S, Giannarelli D, Levato L, Gentile M, Mirabelli R, Morabito F. Do biologic parameters affect the time to first treatment of clinical monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia Rai stage 0? Results of a prospective analysis. Clinical Lymphoma, Myeloma and Leukemia 2015;15(3):e55-60. [DOI] [PubMed] [Google Scholar]
Molica 2019 {published data only}
- Molica S, Giannarelli D, Levato L, Mirabelli R, Levato D, Lentini M, et al. A simple score based on geriatric assessment predicts survival in elderly newly diagnosed chronic lymphocytic leukemia patients. Leukemia & Lymphoma 2019;60(3):845-7. [DOI] [PubMed] [Google Scholar]
Morabito 2011 {published data only}
- Morabito F, De Filippi R, Laurenti L, Zirlik K, Recchia AG, Gentile M, et al. The cumulative amount of serum-free light chain is a strong prognosticator in chronic lymphocytic leukemia. Blood 2011;118(24):6353-61. [DOI] [PubMed] [Google Scholar]
Pepper 2012 {published data only}
- Pepper C, Majid A, Lin TT, Hewamana S, Pratt G, Walewska R, et al. Defining the prognosis of early stage chronic lymphocytic leukaemia patients. British Journal of Haematology 2012;156(4):499-507. [DOI] [PubMed] [Google Scholar]
Qin 2018 {published data only}
- Qin S, Fan L, Liang J, Gale R, Miao Y, Wu Y, et al. Definition of disease-progression risk stratification in untreated chronic lymphocytic leukemia using combined clinical, molecular and virological variables. Hematological Oncology 2018;36(4):656-62. [DOI] [PubMed] [Google Scholar]
Rozman 1982 {published data only}
- Rozman C, Montserrat E, Feliu E, Granena A, Marin P, Nomdedeu B, et al. Prognosis of chronic lymphocytic leukemia: a multivariate survival analysis of 150 cases. Blood 1982;59(5):1001-5. [PubMed] [Google Scholar]
Rozman 1984 {published data only}
- Rozman C, Montserrat E, Rodriguez-Fernandez JM, Ayats R, Vallespi T, Parody R, et al. Bone marrow histologic pattern - the best single prognostic parameter in chronic lymphocytic leukemia: a multivariate survival analysis of 329 cases. Blood 1984;64(3):642-8. [PubMed] [Google Scholar]
Schweighofer 2011 {published data only}
- Schweighofer CD, Coombes KR, Barron LL, Diao L, Newman RJ, Ferrajoli A, et al. A two-gene signature, SKI and SLAMF1, predicts time-to-treatment in previously untreated patients with chronic lymphocytic leukemia. PLOS One 2011;6(12):e28277. [DOI] [PMC free article] [PubMed] [Google Scholar]
Stamatopoulos 2010 {published data only}
- Stamatopoulos B, Meuleman N, De Bruyn C, Pieters K, Anthoine G, Mineur P, et al. A molecular score by quantitative PCR as a new prognostic tool at diagnosis for chronic lymphocytic leukemia patients. PLOS One 2010;5(9):e12780. [DOI] [PMC free article] [PubMed] [Google Scholar]
Tsimberidou 2007 {published data only}
- Tsimberidou AM, Wen S, O'Brien S, McLaughlin P, Wierda WG, Ferrajoli A, et al. Assessment of chronic lymphocytic leukemia and small lymphocytic lymphoma by absolute lymphocyte counts in 2,126 patients: 20 years of experience at the University of Texas M.D. Anderson Cancer Center. Journal of Clinical Oncology 2007;25(29):4648-56. [DOI] [PubMed] [Google Scholar]
Vetro 2018 {published data only}
- Vetro C, Haferlach T, Jeromin S, Stengel A, Zenger M, Nadarajah N, et al. Identification of prognostic parameters in CLL with no abnormalities detected by chromosome banding and FISH analyses. British Journal of Haematology 2018;183(1):47-59. [DOI] [PubMed] [Google Scholar]
Visentin 2015 {published data only}
- Visentin A, Facco M, Frezzato F, Castelli M, Trimarco V, Martini V, et al. Integrated CLL scoring system, a new and simple index to predict time to treatment and overall survival in patients with chronic lymphocytic leukemia. Clinical Lymphoma, Myeloma & Leukemia 2015;15(10):612-20.e5. [DOI] [PubMed] [Google Scholar]
Wierda 2009 {published data only}
- Wierda WG, O'Brien S, Wang X, Faderl S, Ferrajoli A, Do K-A, et al. Characteristics associated with important clinical end points in patients with chronic lymphocytic leukemia at initial treatment. Journal of Clinical Oncology 2009;27(10):1637-43. [DOI] [PMC free article] [PubMed]
References to ongoing studies
NCT00275054 {published data only}
- NCT00275054. Rituximab, fludarabine, and cyclophosphamide or observation alone in treating patients with stage 0, stage I, or stage II chronic lymphocytic leukemia. clinicaltrials.gov/ct2/show/NCT00275054 (first received 11 January 2006).
NCT03436524 {published data only}
- NCT03436524. A prognostic tool for early stage CLL. clinicaltrials.gov/ct2/show/NCT03436524 (first received 19 February 2018).
Additional references
Alba 2017
- Alba AC, Agoritsas T, Walsh M, Hanna S, Iorio A, Devereaux PJ, et al. Discrimination and calibration of clinical prediction models: users guides to the medical literature. JAMA 2017;318(14):1377-84. [DOI] [PubMed] [Google Scholar]
Bouwmeester 2012
- Bouwmeester W, Zuithoff NP, Mallett S, Geerlings MI, Vergouwe Y, Steyerberg EW, et al. Reporting and methods in clinical prediction research: a systematic review. PLOS Medicine 2012;9(5):1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
Cheson 1996
- Cheson BD, Bennett JM, Grever M, Kay N, Keating MJ, O'Brien S, et al. National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood 1996;87(12):4990-7. [PubMed] [Google Scholar]
Chiorazzi 2005
- Chiorazzi N, Rai KR, Ferrarini M. Chronic lymphocytic leukemia. New England Journal of Medicine 2005;352(8):804-15. [PMID: ] [DOI] [PubMed] [Google Scholar]
Collins 2015
- Collins GS, Reitsma JB, Altman DG, Moons KG. Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD). Annals of Internal Medicine 2015;162(1):735-6. [DOI] [PubMed] [Google Scholar]
Collins 2016
- Collins GS, Ma J, Gerry S, Ohuma E, Odondi L, Trivella M, et al. Risk prediction models in perioperative medicine: methodological considerations. Current Anesthesiology Reports 2016;6(3):267-75. [Google Scholar]
Debray 2012
- Debray TP, Koffijberg H, Vergouwe Y, Moons KG, Steyerberg EW. Aggregating published prediction models with individual participant data: a comparison of different approaches. Statistics in Medicine 2012;31(23):2697-712. [DOI] [PubMed] [Google Scholar]
Debray 2014
- Debray TP, Koffijberg H, Nieboer D, Vergouwe Y, Steyerberg EW, Moons KG. Meta-analysis and aggregation of multiple published prediction models. Statistics in Medicine 2014;33(14):2341-62. [DOI] [PubMed] [Google Scholar]
Debray 2017
- Debray TP, Damen JA, Snell KI, Ensor J, Hooft L, Reitsma JB, et al. A guide to systematic review and meta-analysis of prediction model performance. BMJ 2017;356:i6460. [DOI] [PubMed] [Google Scholar]
Debray 2018a
- Debray TP, Damen JA, Riley RD, Snell K, Reitsma JB, Hooft L, et al. A framework for meta-analysis of prediction model studies with binary and time-to-event outcomes. Statistical Methods in Medical Research 2018;28(9):2768-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
Debray 2018b [Computer program]
- metamisc: Diagnostic and prognostic meta-Analysis. R package version 0.1.9. Debray T, Jong V. Vienna, Austria: R Foundation for Statistical Computing, 2018. Available at www.R-project.org.
Dicker 2009
- Dicker F, Herholz H, Schnittger S, Nakao A, Patten N, Wu L, et al. The detection of TP53 mutations in chronic lymphocytic leukemia independently predicts rapid disease progression and is highly correlated with a complex aberrant karyotype. Leukemia 2009;23(1):117-24. [DOI] [PubMed] [Google Scholar]
Döhner 2000
- Döhner H, Stilgenbauer S, Benner A, Leupolt E, Krober A, Bullinger L, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. New England Journal of Medicine 2000;343(26):1910-6. [PMID: ] [DOI] [PubMed] [Google Scholar]
Eichhorst 2015
- Eichhorst B, Robak T, Montserrat E, Ghia P, Hillmen P, Hallek M, et al, ESMO Guidelines Committee. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology 2015;26(Suppl 5):v78-84. [DOI] [PubMed] [Google Scholar]
El Rouby 1993
- El Rouby S, Thomas A, Costin D, Rosenberg CR, Potmesil M, Silber R, et al. p53 gene mutation in B-cell chronic lymphocytic leukemia is associated with drug resistance and is independent of MDR1/MDR3 gene expression. Blood 1993;82(11):3452-9. [PMID: ] [PubMed] [Google Scholar]
Ferrer Lores 2016
- Ferrer Lores B, Navarro Cubells B, Serrano Alcala A, Castillo Martin I, Teruel Casasus A, Ballester Martinez S, et al. Prognostic impact of the new CLL-IPI Index in a single center CLL Spanish cohort. Blood 2016;128(22):2019. [Google Scholar]
Geersing 2012
- Geersing GJ, Bouwmeester W, Zuithoff P, Spijker R, Leeflang M, Moons KG. Search filters for finding prognostic and diagnostic prediction studies in Medline to enhance systematic reviews. PLOS One 2012;7:e32844. [DOI] [PMC free article] [PubMed] [Google Scholar]
Goede 2012
- Goede V, Hallek M. Optimal pharmacotherapeutic management of chronic lymphocytic leukaemia: considerations in the elderly. Drugs and Aging 2012;28(3):163-76. [DOI] [PubMed] [Google Scholar]
Goede 2016
- Goede V, Bahlo J, Kutsch N, Fischer K, Fink AM, Fingerle-Rowson G, et al. Evaluation of the International Prognostic Index for Chronic Lymphocytic Leukemia (CLL-IPI) in elderly patients with comorbidities: analysis of the CLL11 study population. Blood 2016;128(22):4401. [Google Scholar]
Hallek 2008
- Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Döhner H, et al, International Workshop on Chronic Lymphocytic Leukemia. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood 2008;111(12):5446-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
Hallek 2017
- Hallek M. Chronic lymphocytic leukemia: 2017 update on diagnosis, risk stratification, and treatment. American Journal of Hematology 2017;92(9):946-65. [DOI] [PubMed] [Google Scholar]
Herishanu 2013
- Herishanu Y, Katz BZ, Lipsky A, Wiestner A. Biology of chronic lymphocytic leukemia in different microenvironments: clinical and therapeutic implications. Hematology/Oncology Clinics of North America 2013;27(2):173-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
Heus 2018
- Heus P, Damen JA, Pajouheshnia R, Scholten RJ, Reitsma JB, Collins GS, et al. Poor reporting of multivariable prediction model studies: towards a targeted implementation strategy of the TRIPOD statement. BMC Medicine 2018;16(1):120. [DOI] [PMC free article] [PubMed] [Google Scholar]
Howlader 2019
- Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, et al, National Cancer Institute. SEER Cancer Statistics Review, 1975-2016. www.seer.cancer.gov/archive/csr/1975_2016 (based on November 2018 SEER data submission, posted to the SEER web site, April 2019) (accessed May 2019).
Kay 2007
- Kay NE, O'Brien SM, Pettitt AR, Stilgenbauer S. The role of prognostic factors in assessing 'high-risk' subgroups of patients with chronic lymphocytic leukemia. Leukemia 2007;21(9):1885-91. [PMID: ] [DOI] [PubMed] [Google Scholar]
Lefebvre 2019
- Lefebvre C, Glanville J, Briscoe S, Littlewood A, Marshall C, Metzendorf M-I, et al. Chapter 4: Searching for and selecting studies. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.0 (updated July 2019). Cochrane, 2019. Available from www.training.cochrane.org/handbook.
Moher 2009
- Moher D, Liberati A, Tetzlaff J, Altman DG, the PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Journal of Clinical Epidemiology 2009;62(10):1006-12. [DOI] [PubMed] [Google Scholar]
Molica 2018a
- Molica S, Giannarelli D, Mirabelli R, Levato L, Shanafelt TD. Chronic lymphocytic leukemia international prognostic index (CLL-IPI) in patients receiving chemoimmuno or targeted therapy: a systematic review and meta-analysis. Annals of Hematology 2018;97(10):2005-8. [DOI] [PubMed] [Google Scholar]
Molica 2018b
- Molica S, Giannarelli D, Mirabelli R, Levato L, Kay NE, Shanafelt TD. Chronic lymphocytic leukemia international prognostic index: a systematic review and meta-analysis. Blood 2018;131(3):365-8. [DOI] [PubMed] [Google Scholar]
Moons 2009
- Moons KG, Royston P, Vergouwe Y, Grobbee DE, Altman DG. Prognosis and prognostic research: what, why, and how? BMJ 2009;338:b375. [DOI] [PubMed] [Google Scholar]
Moons 2014
- Moons KG, De Groot JA, Bouwmeester W, Vergouwe Y, Mallett S, Altman DG, et al. Critical appraisal and data extraction for systematic reviews of prediction modelling studies: the CHARMS checklist. PLOS Medicine 2014;11:e1001744. [DOI] [PMC free article] [PubMed] [Google Scholar]
Moons 2015
- Moons KG, Altman DG, Reitsma JB, Ioannidis JP, Macaskill P, Steyerberg EW, et al. Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD): explanation and elaboration. Annals of Internal Medicine 2015;162(1):W1-W73. [DOI] [PubMed] [Google Scholar]
Moons 2019
- Moons KG, Wolff RF, Riley RD, Whiting PF, Westwood M, Collins GS, et al. PROBAST: a tool to assess risk of bias and applicability of prediction model studies: explanation and elaboration. Annals of Internal Medicine 2019;170(1):W1-W33. [DOI] [PubMed] [Google Scholar]
Newcombe 2006
- Newcombe RG. Confidence intervals for an effect size measure based on the Mann–Whitney statistic. Part 2: asymptotic methods and evaluation. Statistics in Medicine 2006;25(4):559-73. [DOI] [PubMed] [Google Scholar]
Pajouheshnia 2019
- Pajouheshnia R, Groenwold RH, Peelen LM, Reitsma JB, Moons KG. When and how to use data from randomised trials to develop or validate prognostic models. BMJ 2019;365:l2154. [DOI] [PubMed] [Google Scholar]
Peat 2014
- Peat G, Riley RD, Croft P, Morley KI, Kyzas PA, Moons KG, et al. Improving the transparency of prognosis research: the role of reporting, data sharing, registration, and protocols. PLOS Medicine 2014;11:e1001671. [DOI] [PMC free article] [PubMed] [Google Scholar]
Pflug 2014
- Pflug N, Bahlo J, Shanafelt TD, Eichhorst BF, Bergmann MA, Elter T, et al. Development of a comprehensive prognostic index for patients with chronic lymphocytic leukemia. Blood 2014;124(1):49-62. [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
Riley 2013
- Riley RD, Hayden JA, Steyerberg EW, Moons KG, Abrams K, Kyzas PA, et al. Prognosis research strategy (PROGRESS) 2: Prognostic factor research. PLOS Medicine 2013;10(2):e1001380. [DOI] [PMC free article] [PubMed] [Google Scholar]
Riley 2019
- Riley RD, Van der Windt DA, Croft P, Moons KGM. Prognosis Research in Healthcare. Concepts, Methods, and Impact. Oxford: Oxford University Press, 2019. [Google Scholar]
Shanafelt 2004
- Shanafelt TD, Geyer SM, Kay NE. Prognosis at diagnosis: integrating molecular biologic insights into clinical practice for patients with CLL. Blood 2004;103(4):1202-10. [PMID: ] [DOI] [PubMed] [Google Scholar]
Steyerberg 2013
- Steyerberg EW, Moons KG, Van der Windt DA, Hayden JA, Perel P, Schroter S, et al. Prognosis Research Strategy (PROGRESS) 3: prognostic model research. PLOS Medicine 2013;10(2):e1001381. [DOI] [PMC free article] [PubMed] [Google Scholar]
Steyerberg 2014
- Steyerberg EW, Vergouwe Y. Towards better clinical prediction models: seven steps for development and an ABCD for validation. European Heart Journal 2014;35(29):1925-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
Stilgenbauer 2014
- Stilgenbauer S, Schnaiter A, Paschka P, Zenz T, Rossi M, Döhner K, et al. Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood 2014;123(21):3247-54. [DOI] [PubMed] [Google Scholar]
Van Calster 2019
- Van Calster B, McLernon DJ, Van Smeden M, Wynants L, Steyerberg EW, Bossuyt P, et al. Calibration: the Achilles heel of predictive analytics. BMC Medicine 2019;17(1):230. [DOI] [PMC free article] [PubMed] [Google Scholar]
Viechtbauer 2010
- Viechtbauer W. Conducting meta-analyses in R with the metafor package. Journal of Statistical Software 2010;36(3):1-48. [Google Scholar]
Watson 2008
- Watson L, Wyld P, Catovsky D. Disease burden of chronic lymphocytic leukaemia within the European Union. European Journal of Haematology 2008;81(4):253-8. [DOI] [PubMed] [Google Scholar]
Wierda 2011
- Wierda WG, O'Brien S, Wang X, Faderl S, Ferrajoli A, Do KA, et al. Multivariable model for time to first treatment in patients with chronic lymphocytic leukemia. Journal of Clinical Oncology 2011;29(31):4088-95. [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
Wolff 2019
- Wolff RF, Moons KG, Riley RD, Whiting PF, Westwood M, Collins GS, et al. PROBAST: a tool to assess the risk of bias and applicability of prediction model studies. Annals of Internal Medicine 2019;170(1):51-8. [DOI] [PubMed] [Google Scholar]
Zaja 2013
- Zaja F, Mian M, Volpetti S, Visco C, Sissa C, Nichele I, et al. Bendamustine in chronic lymphocytic leukemia: outcome according to different clinical and biological prognostic factors in the everyday clinical practice. American Journal of Hematology 2013;88(11):955-60. [DOI] [PubMed] [Google Scholar]
Zenz 2011
- Zenz T, Mertens D, Dohner H, Stilgenbauer S. Importance of genetics in chronic lymphocytic leukemia. Blood Reviews 2011;25(3):131-7. [PMID: ] [DOI] [PubMed] [Google Scholar]
References to other published versions of this review
Skoetz 2016
- Skoetz N, Trivella M, Kreuzer KA, Collins G, Köhler N, Wolff M, et al. Prognostic models for chronic lymphocytic leukaemia: an exemplar systematic review and meta‐analysis. Cochrane Database of Systematic Reviews 2016, Issue 1. Art. No: CD012022. [DOI: 10.1002/14651858.CD012022] [DOI] [PMC free article] [PubMed] [Google Scholar]
