Abstract
Durable objective response rate (ORR) remains a meaningful endpoint in recurrent cancer; however, the target ORR for single-arm recurrent glioblastoma trials has not been based on historic information or tied to patient outcomes. The current study reviewed 68 treatment arms comprising 4793 patients in past trials in recurrent glioblastoma in order to judiciously define target ORRs for use in recurrent glioblastoma trials. ORR was estimated at 6.1% [95% CI 4.23; 8.76%] for cytotoxic chemothera + pies (ORR = 7.59% for lomustine, 7.57% for temozolomide, 0.64% for irinotecan, and 5.32% for other agents), 3.37% for biologic agents, 7.97% for (select) immunotherapies, and 26.8% for anti-angiogenic agents. ORRs were significantly correlated with median overall survival (mOS) across chemotherapy (R2= 0.4078, P < .0001), biologics (R2= 0.4003, P = .0003), and immunotherapy trials (R2= 0.8994, P < .0001), but not anti-angiogenic agents (R2= 0, P = .8937). Pooling data from chemotherapy, biologics, and immunotherapy trials, a meta-analysis indicated a strong correlation between ORR and mOS (R2= 0.3900, P < .0001; mOS [weeks] = 1.4xORR + 24.8). Assuming an ineffective cytotoxic (control) therapy has ORR = 7.6%, the average ORR for lomustine and temozolomide trials, a sample size of ≥40 patients with target ORR>25% is needed to demonstrate statistical significance compared to control with a high level of confidence (P < .01) and adequate power (>80%). Given this historic data and potential biases in patient selection, we recommend that well-controlled, single-arm phase II studies in recurrent glioblastoma should have a target ORR >25% (which translates to a median OS of approximately 15 months) and a sample size of ≥40 patients, in order to convincingly demonstrate antitumor activity. Crucially, this response needs to have sufficient durability, which was not addressed in the current study.
Keywords: glioblastoma, objective response rate, overall survival, recurrent GBM
Objective response rate (ORR), defined as the proportion of patients with a specific reduction in tumor size sustained over a predefined minimum amount of time, that is durable is regarded as a valid endpoint for drug approval. According to US FDA guidance, durable ORR can be used as a primary clinical endpoint for traditional approval, a surrogate endpoint for traditional approval, as well as a surrogate endpoint in support of accelerated approval.1 In non-central nervous system (CNS) cancers, the FDA has granted accelerated or even full approval for numerous agents using ORR as a primary endpoint including Abraxane® (paclitaxel protein bound particles) for metastatic breast cancer and non-small cell lung cancer (NSCLC),2 Erivedge® (vismodegib) for basal cell carcinoma,3 Xgeva® (denosumab) for giant cell bone tumors,4 Sutent® (sunitinib malate) for renal cell carcinoma,5 and FOLFOX, or Eloxatin® (oxaliplatin) plus 5-fluorouracil plus leucovorin for metastatic colorectal carcinoma.6 In CNS cancers, everolimus was approved for subependymal giant cell astrocytoma on the basis of a small, single-arm phase 2 trial where a combination of ORR and improvement in symptoms, most notably seizure control were the basis for this approval.7 Importantly, many of these approvals were based on single-arm clinical trials, suggesting this may be a possible path forward for drug development in recurrent glioblastoma (rGBM) if we can succinctly define a meaningful target ORR that will have a strong likelihood of a significant survival benefit. The current manuscript outlines historic evidence supporting the hypothesis that ORR is a meaningful clinical endpoint for most therapeutics as it is strongly related to median overall survival (mOS). In this manuscript, we also outline the target ORR for a given trial size that provides sufficient statistical confidence that the ORR is higher than that of historically non-active therapies often used as a control in rGBM (ie, lomustine).
RANO-Defined ORR
The standard and modified response assessment in neuro-oncology criteria (Response Assessment in Neuro Oncology (RANO)8 and mRANO9), as well as the Macdonald and Levin criteria that preceeded these criteria, define a “response” as having either a “partial response” (PR) or a “complete response” (CR). The ORR is defined as the proportion of all recurrent GBM patients in a clinical trial with measurable disease at baseline (pretreatment) that exhibit a confirmed (durable) PR or CR. All RANO as well as the Macdonald criteria define PR as having ≥50% decrease in the sum of products of perpendicular diameters of all measurable enhancing lesions compared with baseline, and this must be sustained for at least 4 weeks. The first scan exhibiting ≥50% decrease in the sum of products of perpendicular diameters is often considered a “preliminary PR” event, contingent on whether the second scan (made at least 4 weeks later) exhibits a sustained ≥50% decrease in the sum of products with respect to the baseline time point. If the second scan exhibits disease progression (PD) with respect to the “preliminary PR” scan, then the response is not sustained and this is noted as no response. If the second, confirmatory scan exhibits Stable Disease (SD), PR, or CR with respect to the pretreatment baseline, it is considered a durable PR. Importantly, steroid dose is required to be the same or lower compared with the baseline scan and clinical assessments (eg, KPS, ECOG) should also be stable or improved.
Similarly, all RANO criteria as well as the Macdonald criteria consider a CR defined as the disappearance of all enhancing measurable and non-measurable disease, and with RANO this has to be sustained for at least 4 weeks. The first scan exhibiting the disappearance of all enhancing measurable and non-measurable disease is often considered the point of “preliminary CR”, again contingent on whether the second scan (minimum 4 weeks later) confirms the durability of this response. If the second scan continues to exhibit the disappearance of enhancing disease and no emergence of non-measurable disease it is considered a durable CR, dependent on whether the patients are completely off corticosteroids (excluding physiologic replacement doses) and whether the neurological status has stabilized or improved. It is important to note that although confirmation of response is required at a minimum of 4 weeks per RANO recommendations, a more prolonged duration of response (eg, 6 months) provides stronger evidence of treatment efficacy and may be valuable when considering regulatory approval.
RECIST-Defined ORR Benchmarks in Common Solid Tumors
Generally speaking, many solid tumors are vastly more responsive to select therapies compared with GBM. Breast cancers demonstrate Response Evaluation Criteria in Solid Tumors (RECIST)-defined ORRs of between 70 and 80%, as demonstrated by a meta-analysis of 4756 individual patient data from 10 randomized trials in early breast cancer by the Early Breast Cancer Trialists’ Collaborative Group10 and a systematic review and meta-analysis of 2109 patients from 9 randomized controlled trials by Poggio et al10 in triple-negative breast cancer. RECIST-defined ORR in lung cancer is highly dependent on the particular subtype (eg, small cell vs non-small cell, EGFR mutated vs wild type, etc.) and can range from 10 to 70%, with small cell averaging around 60% ORR in first-line and 30–40% during second-line therapy as reported between 1997 and 201711 and roughly 20–40% in NSCLC in phase II and III trials as reviewed article by Shanafelt et al.12 This appears similar to metastatic castration-resistant prostate cancer, which illustrates response rates of 43.5% in rucaparib,13 33–44% in PARP inhibitors,14 and between 12 and 37% in combination immunotherapies.14
Advanced pancreatic cancer (PDA) is similar to GBM in terms of dismal outcomes and lack of treatment options, leading to similarly low ORRs. Similar to GBM, PDA lacks markers for early detection and there are no screening programs and chemotherapy provides only a modest benefit due to the molecular and microenvironmental conditions leading to multiple levels of therapeutic resistance including immune suppression.15,16 As summarized by Lee and Park,17 advanced pancreatic cancer was treated with 5-fluorouracil through the mid-1990s, with ORRs of near zero in phase III trials,18,19 until 1997 when gemcitabine was evaluated in a randomized trial compared with 5-fluorouracil and exhibited an ORR of 5.4%, sustained a minimum of 4 weeks, compared with 0% for 5-fluoruracil. With additional evidence of clinical benefit including a reduction in symptom severity, weight loss, and an increase in survival, gemcitabine was approved as a first-line therapy for advanced pancreatic cancer. Since 1997, trials in advanced pancreatic cancer including gemcitabine combined with a variety of other cytotoxic agents to try and build on the success of gemcitabine monotherapy, including 5-fluorouracil, cisplatin, docetaxel, and irinotecan. Interestingly, this is the same approach that appeared in trials for a time in neuro-oncology, where survival benefits in first-line temozolomide (TMZ) treatment led to a series of relatively unsuccessful trials combining TMZ with other agents. In 2007 the combination of gemcitabine and erlotinib showed a small, yet significant, increase in mOS of 6.3 vs 5.9 months (P = .039) compared to gemcitabine monotherapy in a phase III trial involving 569 patients and this combination was approved for use in first-line advanced pancreatic cancer.20 Importantly, ORR was only 8.6% in the combination compared to 8.0% in gemcitabine monotherapy. In the last decade, more combination trials demonstrated superiority to gemcitabine monotherapy including increased ORR and mOS. Among these trials was a phase III 5-fluorouracil, leucovorin, oxaliplatin, and irinotecan combination (FOLFIRINOX) trial21 in 2011 that showed a response rate of 31.6% compared with gemcitabine monotherapy (9.4%) as well as a near doubling in mOS (11.1 months vs 6.8 months), at the expense of significantly higher toxicity. Additionally, a phase III gemcitabine plus nab-paclitaxel trial22 in 2013 showed a response rate of 23% compared with 7% in the monotherapy arm as well as an improved survival of around 2 months.
When putting historic ORRs for Isocitrate Dehydrogenase (IDH) wild-type recurrent GBM into context with these other solid tumors, it is worth noting that many common solid tumor types have dramatically different ORR and mOS depending on the genetic or phenotypical subtype of cancer, which may or may not have a particularly strong response to targeted therapies (eg, ER+ breast cancer, ALK-positive lung cancer, etc.) or immunotherapies (eg, melanoma, lung cancer subtypes, etc.). Similarly, target response rates and expectations in GBM may need to be adjusted as we identify therapeutically relevant tumor subtypes, although no such subtypes have been identified yet, despite considerable efforts.
Historic Benchmarks for ORR in Recurrent GBM
To establish benchmarks for target ORR in recurrent GBM as well as the association between ORR and mOS, we first searched for representative, recent, later-stage clinical trials (phase II–III when available) in recurrent GBM represented by 4 major therapeutic classifications: cytotoxic chemotherapies, biologic agents (excluding anti-angiogenic agents), anti-angiogenic agents, and immunotherapies. Major studies were included for estimates of ORR if they utilized RANO, Macdonald, Levin, or even the early WHO criteria to define response, but were excluded from examining the correlation between ORR and mOS if they (1) included resective surgery, convection-enhanced delivery (CED), or intratumoral injections as part of the trial, which may bias the results toward longer mOS due to potential cytoreduction of the tumor burden; (2) IDH mutant gliomas included in the trial at a significant proportion (>5–10%), which may similarly lead to a bias in the results toward longer ORR and mOS; (3) if the trial didn’t include the same patients for ORR and mOS evaluations (eg, a subset of patients were evaluated for response based on having a measurable disease, but a larger cohort of patients were evaluated for mOS); and (4) if studies did not explicitly include both ORR and mOS in the published results of the trial. All responses in the included studies were presumed to have a minimum of 4 weeks duration of response as determined by a subsequent confirmatory scan. A search for representative trials resulted in a total of 68 unique treatment arms and 4793 recurrent GBM patients. Meta-analysis was performed in R23 using the metafor package24 applied to published data, including the metaprop() function for estimating ORR and rma() function for estimating the correlation between ORR and mOS. Random effect models were used to account for heterogeneity among the studies in the meta-analysis25–27. We note that a random effects model treats the studies in our meta-analysis as a random sample from an imagined universe of individual studies that might have been performed, which accounts for both between-study and within-study variations. As a result, confidence intervals tend to be wider than that from a fixed-effects meta-analysis. Additionally, simulations to estimate the target ORR for a given sample size (Figure 3) was calculated using PASS28,29 using a one-sided one-proportion test with >80% statistical power.
Figure 3.
Minimum required sample size for (A) a control ORR = 5% (average of all non-antiangiogenic trials); (B) a control ORR = 6.7% (upper 95% confidence interval for all non-antiangiogenic trials); and (C) a control ORR = 7.6% (average from CCNU and TMZ monotherapy trials) assuming a level of significance P < .01 and power >80%. Results suggest a minimum of 40 patients are required for a target ORR of 25% using an assumed control ORR of 7.6%.
Cytotoxic Chemotherapies
A total of 21 study arms from representative trials (Table 1) totaling 1822 recurrent GBM patients were used to estimate the ORR for cytotoxic chemotherapies (Table 1; Figure 1). These treatments were grouped by whether they involved lomustine (CCNU), temozolomide (TMZ), irinotecan (CPT-11), or another type of chemotherapy. Based on these historic data, ORR for CCNU was 7.59% [95% CI 2.87; 18.59%], TMZ was 7.57% [4.75; 11.86%] (including studies with alternative dose scheduling and combinations including TMZ), CPT-11 was 0.64% [0.00; 1.00%], and all other chemotherapies was approximately 5.32% [2.59; 10.62%] (Figure 1; Supplementary Figure S1). These “other chemotherapies” included a summary of data from 437 GBM patients enrolled in 12 different chemotherapy phase II trials within the North American Brain Tumor Consortium between 1998 and 2002 (ORR = 7%),30 the NovoTTF-100A study with 120 patients (ORR = 14%),31 as well as single agent procarbazine (ORR = 5.3%),32 PCV (ORR = 11%),33 hydroxyurea (ORR = 0.8%),34 and combination chemotherapies including BCNU and TMZ (ORR = 5.5%),35 carboplatin and thymidine (ORR = 2.2%),36 and the combination of TMZ, disulfiram, and copper (ORR = 0%).37 ORR across all cytotoxic chemotherapies was approximately 6.1% [4.23; 8.76%] (Figure 1).
Table 1.
Representative Phase ≥ II Trials for Recurrent GBM Patients Evaluating Cytotoxic (Chemotherapy) Agents
| Agent | # Patients | Response criteria | # Recurrences | Primary trial endpoint | ORR (%) | mOS [weeks] |
Citation |
|---|---|---|---|---|---|---|---|
| 12 Phase II NABTC Trials (TMZ, BCNU, CPT-11, etc.) | 437 | Macdonald | ≤3 | PFS6 or ORR | 7 | 30 | Lamborn et al30 |
| Carboplatin + thymidine | 45 | Macdonald | ≤2 | ORR | 2.2 | 23 | Robins et al36 |
| BCNU + TMZ | 36 | Macdonald | ≤1 | PFS6 | 5.5 | 34 | Prados et al35 |
| Carboplatin + erlotinib | 43 | Macdonald | ≤2 | PFS | 2.3 | 30 | de Groot et al38 |
| CCNU | 92 | Levin | ≤2 | PFS | 4.3 | 28.6 | Wick et al39 |
| CCNU | 65 | RANO | 1 | PFS | 8.9 | 42.5 | Batchelor et al40 |
| CCNU | 137 | RANO | 1 | OS | 13.9 | 37.4 | Wick et al41 |
| CCNU (BELOB) | 46 | RANO | 1 | OS9 | 5 | 34.8 | Taal et al42 |
| Hydroxyurea | 120 | Macdonald | 1 | PFS | 0.8 | 19 | Dresemann et al34 |
| CPT-11 | 40 | Macdonald | ≤2 | ORR | 0 | 22.2 | Santisteban et al43 |
| CPT-11 | 48 | Macdonald | ≤1 | ORR | 17 | 43 | Friedman et al44 |
| CPT-11 | 40 | Macdonald | ≤1 | ORR | 0 | 17.4 | Chamberlain et al45 |
| PCV | 63 | Macdonald | ≤1 | ORR | 11 | 33 | Kappelle et al33 |
| Procarbazine | 113 | Macdonald | ≤1 | PFS6 | 5.3 | 25.6 | Yung et al32 |
| TMZ | 112 | Macdonald | ≤1 | PFS6 | 5.4 | 27.8 | Yung et al32 |
| TMZ | 128 | Macdonald | 1 | PFS6 | 8 | 23.5 | Brada et al46 |
| TMZ* | 33 | Macdonald | 1 | PFS6 | 9 | 40 | Brandes et al47 |
| TMZ* | 54 | Macdonald | 1 | PFS6 | 13 | 51.3 | Norden et al48 |
| TMZ* | 27 | RECIST | 1 | PFS6 | 11.1 | 40.4 | Perry et al49 |
| TMZ + disulfiram + Cu | 23 | RANO | 1 | ORR | 0 | 30.9 | Huang et al37 |
| NovoTTF-100A | 120 | Macdonald | No limit | OS | 14 | 28.7 | Stupp et al31 |
TMZ = Temozolomide.
TMZ* = Temozolomide with alternative dose scheduling.
CPT-11 = Irinotecan.
BCNU = Carmustine.
CCNU = Lomustine.
Cu = Copper.
RANO = Response assessment in neuro-oncology criteria.
PCV = Procarbazine hydrochloride, lomustine, and vincristine sulfate.
PFS = Progression-free survival.
PFS6 = Proportion of patients with PFS at or beyond 6 months from start of treatment (or randomization).
OS = Overall survival.
OS9 = Proportion of patients alive after 9 months from the start of treatment (or randomization).
Figure 1.
Objective response rates (ORRs) for various types of treatments including cytotoxic chemotherapies, biologic-based agents, immunotherapies (select trials), and anti-angiogenic agents. Filled circle under biologic-based agents shows the ORR for a recent trial of dabrafenib and trametinib in BRAF-mutated rGBM. Filled square under “chemotherapies” designates the ORR for NovoTTF-11A.
Biologic-Based (Non-Angiogenic) Therapies
A total of 20 treatment arms from 18 representative studies (Table 2) totaling 1249 recurrent GBM patients were used to estimate ORR for biologic-based (non-angiogenic) therapies. While the targets and penetration of these agents vary widely, these data suggest that many of these agents have little efficacy in terms of radiographic response, with one exception. While studies evaluating rilotumumab,50 erlotinib plus sirolimus,51 fenretinib,52 temsirolimus,53 perifosine,54 and buparlisib55 failed to demonstrate a single patient with a radiographic response, one recent and notable study by Wen et al56 demonstrated an ORR of 32% in BRAF mutant rGBM patients when treated with dabrafenib and trametinib. Of note, dabrafenib and trametinib recently received accelerated FDA approval in BRAF V600E mutant solid tumors including rGBM. The ORR across all treatment arms evaluated excluding BRAF mutant rGBM was 3.37% [1.81; 6.18%] (Figure 1; Supplemental Figure S2A). However, because of the broad range of targets, penetration, and evaluation criteria, it is challenging to generalize about efficacy across this broad category of therapeutics.
Table 2.
Representative Recent Phase ≥ II Clinical Trials for Recurrent GBM Patients Evaluating Non-angiogenic, Biologic-Based Therapeutics
| Agent | # Patients | Response Criteria |
# Recurrences | Primary trial endpoint | ORR (%) | mOS [Weeks] |
Citation |
|---|---|---|---|---|---|---|---|
| Rilotumumab | 61 | Macdonald | ≤3 | ORR | 0 | 23.5 | Wen et al50 |
| Cilengitide | 81 | Macdonald | ≤1 | PFS6 | 9 | 43 | Reardon et al57 |
| Enzastaurin | 174 | Levin | ≤2 | PFS | 2.9 | 28.7 | Wick et al39 |
| Erlotinib | 54 | Macdonald | ≤1 | PFS6 | 3.7 | 33.5 | van den Bent et al58 |
| Erlotinib | 48 | WHO | ≤1 | ORR | 6.3 | 42.1 | Yung et al59 |
| Erlotinib + sirolimus | 32 | Macdonald | 1 | PFS6 | 0 | 33.8 | Reardon et al51 |
| Fenretinib | 23 | Not stated | ≤2 | PFS6 | 0 | 30 | Puduvalli et al52 |
| Imatinib | 51 | Macdonald | ≤2 | ORR or PFS6 | 5.9 | 23 | Raymond et al60 |
| Imatinib + hydroxyurea | 33 | Macdonald | No limit | PFS6 | 9 | 48.9 | Reardon et al61 |
| Imatinib + hydroxyurea | 231 | Macdonald | 1 | ORR | 3.4 | 26 | Reardon et al62 |
| Imatinib + hydroxyurea | 120 | Macdonald | 1 | PFS | 1.7 | 21 | Dresemann et al34 |
| Temsirolimus | 65 | Macdonald | ≤2 | PFS6 | 0 | 19 | Galanis et al53 |
| Vorinostat | 66 | Macdonald | ≤2 | ORR | 3.0 | 24.8 | Galanis et al43 |
| Perifosine | 16 | RANO | No limit | PFS6 | 0 | 14.7 | Kaley et al54 |
| Buparlisib | 65 | RANO | ≤2 | PFS6 | 0 | 42.6 | Wen et al55 |
| Marizomib | 30 | RANO | ≤2 | ORR | 3.3 | 39.5 | Bota et al63 |
| Selinexor | 68 (24 50 mg 2xWK) (14, 60 mg 2xWK) (30, 80 mg 1xWK) |
RANO | ≤3 | PFS6 | 8.3 (50 mg) 7.7 (60 mg) 10 (80 mg) |
45.6 33.5 43.5 |
Lassman et al64 |
| † Dabrafenib + trametinib (in BRAF mutant rGBM)* | 31 | RANO | 1 | ORR | 32 | 59.5 | Wen et al56 |
†Believed to be a “positive” study, with ORR = 32% and mOS of more than a year.
*Note this involved an enriched patient population.
PFS = Progression-free survival.
PFS6 = Proportion of patients with PFS at or beyond 6 months from start of treatment (or randomization).
OS = Overall survival.
Immunotherapies
Accurately estimating ORRs for recurrent GBM treated with immunotherapies was similarly challenging, as there are a variety of different therapeutic targets, delivery methods, and many studies that enrich for particular subpopulations of patients (eg, PD-L1 positive, etc.). Many immunotherapy trials involve either a significant surgery (eg, to obtain enough tissue for a vaccine) or surgical procedure (eg, CED, intratumoral injections), or trials reported ORR in only a subset of patients with measurable disease and mOS for the entire treatment cohort.65 Additionally, some studies utilized RECIST criteria (eg, Reardon et al66 and Izumoto et al67), so these studies were not included in the final estimates of ORR or the relationship between ORR and mOS. In the end, a total of 11 treatment arms from 10 trials totaling N = 436 recurrent GBM patients were included in estimates of ORR (Supplementary Table S1). Among these important studies are those involving single agent nivolumab65,68 (ORR = 7.8–11%), pembrolizumab69 (ORR = 0%), the combination of nivolumab and ipilimumab68 (ORR = 10%), as well as oncolytic viral therapies70–72 (ORR = 4.7–13%). The ORR across all these immunotherapy studies was 7.97% [5.32; 11.77%] (Figure 1; Supplementary Figure S2B).
The ORR across all non-anti-angiogenic agent studies, including cytotoxic therapies, biologics, and immunotherapies was 5.03% [3.76; 6.69].
Anti-Angiogenic Agents
Contrast enhancement on post-contrast T1-weighted MR images serves as a reasonable surrogate for tumor burden in GBM.73 However, contrast enhancement relies on extravasation of intravascular contrast into the extravascular space as a consequence of increased vascular permeability in regions of neovasculature. Anti-angiogenic agents, which mostly reflect anti-VEGF agents and drugs that target tumor vasculature, by nature may alter vascular characteristics including permeability and have demonstrated a significant reduction in the extent and intensity of contrast enhancement,74–76 resulting in artificially high ORRs in rGBM for this class of therapeutics when compared with other types of treatment. A total of 16 treatment arms from 10 trials including N = 1286 rGBM patients were included (Supplementary Table S2). The ORR for all studies involving bevacizumab,42,65,75,77,78 single agent or in combination, was around 28.19% [23.02; 34.01%] while the ORR for all study arms examined was 26.8% [21.59; 32.66%], (Figure 1; Supplementary Figure S2C). Importantly, the trials included in this list were those known to have significant effects on contrast-enhanced MRI. However, pazopanib, sorafenib, regorafenib, and other agents that do not demonstrate such strong changes in vascular permeability may require additional consideration.
Association Between ORR and mOS in rGBM
Next, the potential association between ORR and mOS was explored for each of the treatment groups separately. When examining relevant cytotoxic chemotherapy trials (Table 1), a strong linear correlation was observed between ORR and mOS (Figure 2A; R2= 0.4078, P < .0001). Similarly, examination of biologic-based therapeutics, excluding the study by Wen et al56 showed a strong linear correlation (Figure 2B; R2= 0.4003, P = .0003). When the study by Wen et al56 was included, which represents a recent study contributing to accelerated FDA approval of dabrafenib and trematinib, this association was stronger (R2= 0.5024, P < .0001, results not shown).
Figure 2.
Correlation between ORR and median overall survival (mOS) for (A) cytotoxic chemotherapies, (B) biologics-based (non-antiangiogenic) agents, (C) immunotherapies (select studies), and (D) anti-angiogenic agents. Note that a strong correlation was observed in all therapeutic categories besides anti-angiogenic agents. (E) Correlation between ORR and mOS for pooled studies in chemotherapies, biologics-based agents, and immunotherapies (46 treatment arms in 3243 rGBM patients). ORR vs mOS in pooled patients split by individual treatment types. showing strong linear correlation (R2= 0.3900, P < .0001; mOS [weeks] = 1.4xORR + 24.8). (F) ORR vs mOS pooled together along with ORR and mOS in recent study in a recent trial of dabrafenib and trametinib in BRAF-mutated rGBM (filled circle).
Evaluation of the association between ORR and mOS was more challenging for immunotherapy trials investigated, as studies needed to be excluded if they (1) only reported ORR for a subset of patients, but reported mOS for the larger cohort; 65 (2) used the RECIST criteria instead of the WHO, Macdonald, RANO, or mRANO criteria,66,67 which have more similar definitions of response; (3) included a relatively large proportion of IDH mutant tumors, potentially skewing mOS estimates; 69 and (4) involved surgical resection of the tumor or CED of study drug.72,79 This resulted in a total of 5 treatment arms from 4 trials in only N = 138 patients. However, a linear correlation was also observed in this limited dataset of immunotherapy trials (Figure 2C; R2= 0.8994, P < .0001). (Notably, this association was based on a very limited number of trials and small sample sizes, so these results should be interpreted with caution).
Lastly, the association between ORR and mOS was explored in anti-angiogenic therapies (Figure 2D). No significant association was observed between ORR and mOS for this class of agents, either as single agents or in combination (Figure 2D; R2= 0, P = .8937).
Pooling together the patients included in the chemotherapy, biologics-based therapies, and select immunotherapy trials, excluding the study by Wen et al56 resulted in 46 treatment arms from a combined N = 3243 rGBM patients available to generalize the association between ORR and mOS. A strong, positive, linear correlation was observed between ORR and mOS (Figure 2E; R2= 0.3900, P < .0001), with a slope of 1.4 ± 0.3 and an intercept of 24.8 ± 1.9 weeks. This results in the linear equation mOS[weeks] = 1.4xORR + 24.8 allowing us to extrapolate and estimate the resulting mOS for given ORR thresholds. For example, an ORR>25% would result in an approximate mOS of 15 months (60 weeks), around double the mOS expected from an ORR = 5% (31.8 weeks, average of all non-anti-angiogenic trials), while an ORR of >40% would result in an approximate mOS of 20 months (80.8 weeks). When the study by Wen et al56 is superimposed on this data, representing a study that resulted in accelerated approval at least in part due to the high ORR, we can clearly see that the high ORR appears to be consistent with, but slightly lower than, the projected estimate of mOS (Figure 2F). Together, this data supports the use of the general association between ORR and mOS to predict the desired survival benefit for given the target ORR.
Estimating Target ORR for a Given Study Sample Size
While an ORR of 25% appears to be an appropriately high bar to essentially double the mOS when compared with relatively ineffective therapies, the choice of the target ORR for a given trial should be considered within the context of statistical power for a given study sample size, while also maintaining a minimum number of patients to ensure there is no skew from potential bias in accrual. If we assume that ineffective therapies have an ORR of 5%, or the average ORR from cytotoxic, biologics, and select immunotherapy trials, then ≥26 patients are required for a target ORR of >25% (≥7 of 26) in order to demonstrate statistical significance with a high level of confidence (P < .01) and adequate statistical power (1-β>80%) (Figure 3A) assuming a one-sided interval for one proportion using the Clopper-Pearson method for estimating the confidence intervals.80 Alternatively, one might consider a more cautious assumption for the control arm ORR based on the upper bound of the 95% confidence interval for the distribution of ORR across all non-anti-angiogenic trials, or an ORR = 6.7%. Using this assumed ORR for the control group, ≥35 patients are required for a target ORR of >25% (≥9 of 35) in order to demonstrate statistical significance (P < .01) with power >80% (Figure 3B). An even more conservative, and potentially more realistic, estimate for ORR for the control arm would be to use an ORR based on the average ORR observed in lomustine and temozolomide studies, or ORR = 7.6%, as lomustine is often used as the control arm in rGBM trials39–42 including contemporary trials such as GBM AGILE.81 Using an assumed control ORR of 7.6%, ≥40 patients are required for a target ORR of >25% (>10 of 40) in order to demonstrate statistical significance (P < .01) with power >80% (Figure 3C). It is important to note that more than 40 patients may be required to ensure adequate confidence in the results, particularly if pretreatment tumor growth is not confirmed before entering the trial82,83 and if tumors are relatively small,84 as these factors can skew the number of observed responses and significantly impact outcomes.85 In contrast, the use of a standardized brain tumor imaging protocol86 and standardized approaches to central radiographic reads87 may help reduce noise in our estimate of ORR. Thus, we recommend rGBM studies contain a minimum sample size of 40 and strive for a target ORR >25% (>10 of 40), while adhering to standardized image acquisition, radiographic read paradigm, and response assessment guidance.
Conclusions and Limitations
Durable ORR remains an important endpoint in a number of cancers, including recurrent GBM. After reviewing contemporary studies, the median ORR was estimated to be around 6.1% for cytotoxic chemotherapies (7.6% for lomustine and temozolomide), 3.37% for biologic agents, 7.97% for select immunotherapy trials, and 26.8% for anti-angiogenic agents. While ORRs were significantly correlated with median overall survival (mOS) across chemotherapy, biologics, and immunotherapy trials, we did not find a strong correlation between ORR and mOS in anti-angiogenic agents that are known to reduce blood-brain barrier permeability and alter the degree of contrast enhancement. Combined data from chemotherapy, biologics, and immunotherapy trials showed a strong correlation between ORR and mOS, and the trendline describing this relationship suggests an ORR>25% results in an mOS of more than 15 months. Finally, it should be noted that we do not have comparative data in prospective randomized studies on recurrent glioblastoma, confirming changes in ORR translate in OS benefit. While the use of ORR as a primary endpoint in single-arm studies is not as controversial as the use of time-to-event endpoints including PFS and OS, prospective corroboration of the observed trends in a randomized trial would strengthen the claims made in the current manuscript.
It is important to point out a few limitations and confounds to the current study. First, we attempted to report and include ORRs from a wide variety of studies that utilized RANO, Macdonald, and Levin, which all have similar, but not equivalent, definitions of response and requirements for durability of response (DOR). Additionally, the association between ORR and mOS for immunotherapy trials should be interpreted with caution because it was based on a very limited number of trials and small sample sizes, as many studies were excluded for a variety of reasons including a mismatch between the patients included in ORR and mOS estimates. Additionally, we did not have access to patient-level data for our analyses, so questions surrounding the impact of known prognostic variables (eg, MGMT status, etc.) were not specifically addressed in the current study, which may have led to some variability in ORRs across the same drug or drug category. Consequently, only study-level, published data was included in the current meta-analysis. Therefore, it is important to point out that publication bias, data availability bias, and reviewer selection biases are of potential concern when interpreting our results.88–92 Indeed, our estimation of publication bias and data heterogeneity support this concern (see Supplementary Data). Another potential limitation is uncertainty around the proportion of patients who truly exhibited progressive disease at the time of trial entry, as patients with unrecognized pseudoprogression could contaminate our estimates of true therapeutic response. Lastly, and perhaps most critically, radiographic responses need to have sufficient durability, which was not directly addressed in the current study. While most of the studies included required confirmation of response >4 weeks after response was first observed in order to be considered a response, durable responses much longer than this are almost certainly required to truly make a real impact on survival this disease. While the >4 week confirmation scan sets the minimum durability that appears meaningful in recurrent glioblastoma, except for in the setting of potent anti-VEGF therapies, it is likely the both a high ORR and DOR will be required for prolonged survival.
In summary, we recommend a minimum target ORR for phase II studies in recurrent GBM of more than 25%, for a sample size of at least 40 patients, in order to reach a target mOS of around 15 months and ensure the ORR is higher than 7.6%, the average ORR for historic lomustine and temozolomide trials. Radiographic responses should be determined according to RANO criteria, including the use of a confirmation scan a minimum of 4 weeks after the initial response. Importantly, the framework we have laid out for rGBM can also be used for other tumor types, including IDH mutant tumors and diffuse midline gliomas.
Supplementary Material
Contributor Information
Benjamin M Ellingson, UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, Los Angeles, California, USA; UCLA Neuro-Oncology Program, Los Angeles, California, USA; Department of Radiological Sciences, Los Angeles, California, USA; Department of Psychiatry and Biobehavioral Sciences, Los Angeles, California, USA; Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
Patrick Y Wen, Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA.
Susan M Chang, Division of Neuro-Oncology, University of California San Francisco, San Francisco, California, USA.
Martin van den Bent, Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands.
Michael A Vogelbaum, Department of NeuroOncology, Moffitt Cancer Center, Tampa, Florida, USA.
Gang Li, Department of Biostatistics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
Shanpeng Li, Department of Biostatistics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
Jiyoon Kim, Department of Biostatistics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
Gilbert Youssef, Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA.
Wolfgang Wick, Neurology Clinic, University of Heidelberg and Clinical Cooperation Unit Neuro-oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Andrew B Lassman, Division of Neuro-Oncology, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, New York-Presbyterian Hospital, New York, New York, USA.
Mark R Gilbert, Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA.
John F de Groot, Division of Neuro-Oncology, University of California San Francisco, San Francisco, California, USA.
Michael Weller, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland.
Evanthia Galanis, Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA.
Timothy F Cloughesy, UCLA Neuro-Oncology Program, Los Angeles, California, USA; Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
Funding
There was no study-specific funding. Outside this work, funding was provided by the National Brain Tumor Society (NBTS), the UCLA SPORE in Brain Cancer (NIH/NCI P50 CA211015) and the Harvard SPORE in Brain Cancer (NIH/NCI P50 CA165962).
Conflict of Interest
BME is a Paid Consultant and Advisor for Siemens; Medicenna; MedQIA; Imaging Endpoints; Agios Pharmaceuticals; Neosoma; Janssen; Kazia; VBL; Oncoceutics; Boston Biomedical Inc; ImmunoGenesis; and Ellipses Pharma. BME has received grant funding from Siemens, Agios, and Janssen. T.F.C. is cofounder, major stock holder, consultant and board member of Katmai Pharmaceuticals, member of the board for the 501c3 Global Coalition for Adaptive Research, holds stock option of Notable Labs, holds stock in Chimerix and receives milestone payments and possible future royalties, member of the scientific advisory board for Break Through Cancer, member of the scientific advisory board for Cure Brain Cancer Foundation, has provided paid consulting services to GCAR; Gan & Lee; BrainStorm; Katmai; Sapience; Inovio; Vigeo Therapeutics; DNATrix; Tyme; SDP; Novartis; Roche; Kintara; Bayer; Merck; Boehinger Ingelheim; VBL; Amgen; Kiyatec; Odonate Therapeutics QED; Medefield; Pascal Biosciences; Bayer; Tocagen; Karyopharm; GW Pharma; Abbvie; VBI; Deciphera; VBL; Agios; Genocea; Celgene; Puma; Lilly; BMS; Cortice; Wellcome Trust; Novocure; Novogen; Boston Biomedical; Sunovion; Human Longevity; Insys; ProNai; Pfizer; Notable labs; Medqia Trizel; Medscape and has contracts with UCLA for the Brain Tumor Program with Oncovir; Merck; Oncoceutics; Novartis; Amgen; Abbvie; DNAtrix; Beigene; BMS; AstraZeneca; Kazia; Agios; Boston Biomedical; Deciphera; Tocagen; Orbus; AstraZenica; Karyopharm. M.J.v.d.B. has received honoraria from Carthera; Genenta; Nerviano; Cellgene; Astra Zeneca and Boehringer. H.C. has received in the last year honoraria or consulting fees from Best Doctors/Teladoc; Orbus Therapeutics; Adastra Pharmaceuticals; Bristol Meyers Squibb and research funding (instutional contracts) from Orbus; Merck; DNATrix; Abbvie; Beigene; Forma Therapeutics; GCAR; Array BioPharma; Karyopharm Therapeutics; Nuvation Bio; Bayer; Bristol Meyer Squib. W.W. reports to be inventor and patent-holder on “Peptides for use in treating or diagnosing IDH1R132H positive cancers” (EP2800580B1) and “Cancer therapy with an oncolytic virus combined with a checkpoint inhibitor” (US11027013B2). He consulted for Apogenix; Astra Zeneca; Bayer; Enterome; Medac; MSD; and Roche/Genentech with honoraria paid to the Medical Faculty at the University of Heidelberg. MW has received research grants from Apogenix and Quercis, and honoraria for lectures or advisory board participation or consulting from Bayer; Medac; Merck (EMD); Nerviano Medical Sciences; Novartis; Orbus; Philogen and y-Mabs. MAV has patent royalty rights from Infuseon Therapeutics. He has received honoraria from Olympus and Chimerix. His institution has received grant funding from Infuseon Therapeutics; Oncosynergy; and Denovo Pharma for clinical trials for which he is the site primary investigator. EG has received honoraria for advisory board participation from Kiyatec, Inc. (personal compensation) and Karyopharm Therapetuics, Inc. for Data Safety and Monitoring Board participation (compensation to employer). Her institution has received grant funding from Servier Pharmaceuticals LLC (formerly Agios Pharmaceuticals, Inc.), Celgene, MedImmune, Inc. and Tracon Pharmaceuticals.
Author Contribution
Drs. Ellingson, Wen and Cloughesy conceived the review. All authors took part in discussions, analysis, editing and final approval of the manuscript.
References
- 1. U.S. Department of Health and Human Services, Food and Drug Administration, Oncology Center for Excellence, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER). Clinical Trial Endpoints for the Approval of Cancer Drugs and Biologics: Guidence for Industry; 2018. Silver Spring, MD; U.S. DHHS FDA. https://www.fda.gov/media/71195/download. Accessed November 1, 2022. [Google Scholar]
- 2. Ma P, Mumper RJ. Paclitaxel nano-delivery systems: a comprehensive review. J Nanomed Nanotechnol. 2013; 4(2):1000164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Axelson M, Liu K, Jiang X, et al. U.S. food and drug administration approval: vismodegib for recurrent, locally advanced, or metastatic basal cell carcinoma. Clin Cancer Res. 2013; 19(9):2289–2293. [DOI] [PubMed] [Google Scholar]
- 4. Federman N, Brien EW, Narasimhan V, et al. Giant cell tumor of bone in childhood: clinical aspects and novel therapeutic targets. Paediatr Drugs. 2014; 16(1):21–28. [DOI] [PubMed] [Google Scholar]
- 5. Wood L. Sunitinib malate for the treatment of renal cell carcinoma. Expert Opin Pharmacother. 2012; 13(9):1323–1336. [DOI] [PubMed] [Google Scholar]
- 6. Ibrahim A, Hirschfeld S, Cohen MH, et al. FDA drug approval summaries: oxaliplatin. Oncologist. 2004; 9(1):8–12. [DOI] [PubMed] [Google Scholar]
- 7. Krueger DA, Care MM, Holland K, et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med. 2010; 363(19):1801–1811. [DOI] [PubMed] [Google Scholar]
- 8. Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 10 2010; 28(11):1963–1972. [DOI] [PubMed] [Google Scholar]
- 9. Ellingson BM, Wen PY, Cloughesy TF. Modified criteria for radiographic response assessment in glioblastoma clinical trials. Neurotherapeutics. 2017; 14(2):307–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Poggio F, Bruzzone M, Ceppi M, et al. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis. Ann Oncol. 2018; 29(7):1497–1508. [DOI] [PubMed] [Google Scholar]
- 11. Lattuca-Truc M, Timsit JF, Levra MG, et al. Trends in response rate and survival in small-cell lung cancer patients between 1997 and 2017. Lung Cancer. 2019; 131:122–127. [DOI] [PubMed] [Google Scholar]
- 12. Shanafelt TD, Loprinzi C, Marks R, Novotny P, Sloan J. Are chemotherapy response rates related to treatment-induced survival prolongations in patients with advanced cancer? J Clin Oncol. 2004; 22(10):1966–1974. [DOI] [PubMed] [Google Scholar]
- 13. Abida W, Patnaik A, Campbell D, et al. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a brca1 or brca2 gene alteration. J Clin Oncol. 2020; 38(32):3763–3772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Powers E, Karachaliou GS, Kao C, et al. Novel therapies are changing treatment paradigms in metastatic prostate cancer. J Hematol Oncol. 2020; 13(1):144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Oberstein PE, Olive KP. Pancreatic cancer: why is it so hard to treat? Therap Adv Gastroenterol. 2013; 6(4):321–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Zhu H, Li T, Du Y, Li M. Pancreatic cancer: challenges and opportunities. BMC Med. 2018; 16(1):214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Lee HS, Park SW. Systemic chemotherapy in advanced pancreatic cancer. J Chest Surg. 2016; 10(3):340–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. DeCaprio JA, Mayer RJ, Gonin R, Arbuck SG. Fluorouracil and high-dose leucovorin in previously untreated patients with advanced adenocarcinoma of the pancreas: results of a phase II trial. J Clin Oncol. 1991; 9(12):2128–2133. [DOI] [PubMed] [Google Scholar]
- 19. Van Rijswijk RE, Jeziorski K, Wagener DJ, et al. Weekly high-dose 5-fluorouracil and folinic acid in metastatic pancreatic carcinoma: a phase II study of the EORTC GastroIntestinal Tract Cancer Cooperative Group. Eur J Cancer. 2004; 40(14):2077–2081. [DOI] [PubMed] [Google Scholar]
- 20. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the national cancer institute of Canada clinical trials group. J Clin Oncol. 2007; 25(15):1960–1966. [DOI] [PubMed] [Google Scholar]
- 21. Conroy T, Desseigne F, Ychou M, et al. Folfirinox versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011; 364(19):1817–1825. [DOI] [PubMed] [Google Scholar]
- 22. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013; 369(18):1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Balduzzi S, Rücker G, Schwarzer G. How to perform a meta-analysis with R: a practical tutorial. Evid Based Ment Health. 2019; 22(4):153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Viechtbauer W. Conducting meta-analyses in R with the metafor Package. J Stat Softw. 2010; 36(3):1–48. [Google Scholar]
- 25. DerSimonian R, Kacker R. Random-effects model for meta-analysis of clinical trials: an update. Contemp Clin Trials. 2007; 28(2):105–114. [DOI] [PubMed] [Google Scholar]
- 26. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986; 7(3):177–188. [DOI] [PubMed] [Google Scholar]
- 27. Berkey CS, Hoaglin DC, Mosteller F, Colditz GA. A random-effects regression model for meta-analysis. Stat Med. 1995; 14(4):395–411. [DOI] [PubMed] [Google Scholar]
- 28. Fleiss JL, Levin, B, Paik, M.C.. Statistical Methods f or Rates and Proportions. 3rd ed 2003. New Jersey, John Wiley & Sons, Inc. [Google Scholar]
- 29. Newcombe RG. Two-sided confidence intervals for the single proportion: comparison of seven methods. Stat Med. 1998; 17(8):857–872. [DOI] [PubMed] [Google Scholar]
- 30. Lamborn KR, Yung WK, Chang SM, et al. Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol. 2008; 10(2):162–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Stupp R, Wong ET, Kanner AA, et al. NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer. 2012; 48(14):2192–2202. [DOI] [PubMed] [Google Scholar]
- 32. Yung WK, Albright RE, Olson J, et al. A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br J Cancer. 2000; 83(5):588–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Kappelle AC, Postma TJ, Taphoorn MJ, et al. PCV chemotherapy for recurrent glioblastoma multiforme. Neurology. 2001; 56(1):118–120. [DOI] [PubMed] [Google Scholar]
- 34. Dresemann G, Weller M, Rosenthal MA, et al. Imatinib in combination with hydroxyurea versus hydroxyurea alone as oral therapy in patients with progressive pretreated glioblastoma resistant to standard dose temozolomide. J Neurooncol. 2010; 96(3):393–402. [DOI] [PubMed] [Google Scholar]
- 35. Prados MD, Yung WA, Fine HA, et al. Phase 2 study of BCNU and temozolomide for recurrent glioblastoma multiforme: North American Brain Tumor Consortium study. Neuro Oncol. 2004; 6(1):33–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Robins HI, Chang SM, Prados MD, et al. A phase II trial of thymidine and carboplatin for recurrent malignant glioma: a North American Brain Tumor Consortium Study. Neuro Oncol. 2002; 4(2):109–114. [PMC free article] [PubMed] [Google Scholar]
- 37. Huang J, Chaudhary R, Cohen AL, et al. A multicenter phase II study of temozolomide plus disulfiram and copper for recurrent temozolomide-resistant glioblastoma. J Neuro Oncol. 2019; 142(3):537–544. [DOI] [PubMed] [Google Scholar]
- 38. de Groot JF, Gilbert MR, Aldape K, et al. Phase II study of carboplatin and erlotinib (Tarceva, OSI-774) in patients with recurrent glioblastoma. J Neurooncol. 2008; 90(1):89–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Wick W, Puduvalli VK, Chamberlain MC, et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol. 2010; 28(7):1168–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Batchelor TT, Mulholland P, Neyns B, et al. Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J Clin Oncol. 2013; 31(26):3212–3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Wick W, Gorlia T, Bendszus M, et al. Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017; 377(20):1954–1963. [DOI] [PubMed] [Google Scholar]
- 42. Taal W, Oosterkamp HM, Walenkamp AM, et al. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014; 15(9):943–953. [DOI] [PubMed] [Google Scholar]
- 43. Santisteban M, Buckner JC, Reid JM, et al. Phase II trial of two different irinotecan schedules with pharmacokinetic analysis in patients with recurrent glioma: North Central Cancer Treatment Group results. J Neurooncol. 2009; 92(2):165–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Friedman HS, Petros WP, Friedman AH, et al. Irinotecan therapy in adults with recurrent or progressive malignant glioma. J Clin Oncol. 1999; 17(5):1516–1516. [DOI] [PubMed] [Google Scholar]
- 45. Chamberlain MC. Salvage chemotherapy with CPT-11 for recurrent glioblastoma multiforme. J Neurooncol. 2002; 56(2):183–188. [DOI] [PubMed] [Google Scholar]
- 46. Brada M, Hoang-Xuan K, Rampling R, et al. Multicenter phase II trial of temozolomide in patients with glioblastoma multiforme at first relapse. Ann Oncol. 2001; 12(2):259–266. [DOI] [PubMed] [Google Scholar]
- 47. Brandes AA, Tosoni A, Cavallo G, et al. Temozolomide 3 weeks on and 1 week off as first-line therapy for recurrent glioblastoma: phase II study from gruppo italiano cooperativo di neuro-oncologia (GICNO) Br J Cancer. 2006; 95(9):1155–1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Norden AD, Lesser GJ, Drappatz J, et al. Phase 2 study of dose-intense temozolomide in recurrent glioblastoma. Neuro Oncol. 2013; 15(7):930–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Perry JR, Bélanger K, Mason WP, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol. 2010; 28(12):2051–2057. [DOI] [PubMed] [Google Scholar]
- 50. Wen PY, Schiff D, Cloughesy TF, et al. A phase II study evaluating the efficacy and safety of AMG 102 (rilotumumab) in patients with recurrent glioblastoma. Neuro Oncol. 2011; 13(4):437–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Reardon DA, Desjardins A, Vredenburgh JJ, et al. Phase 2 trial of erlotinib plus sirolimus in adults with recurrent glioblastoma. J Neurooncol. 2010; 96(2):219–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Puduvalli VK, Yung WKA, Hess KR, et al. Phase II study of fenretinide (NSC 374551) in adults with recurrent malignant gliomas: a North American Brain Tumor Consortium Study. J Clin Oncol. 2004; 22(21):4282–4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Galanis E, Buckner JC, Maurer MJ, et al. Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a North Central Cancer Treatment Group Study. J Clin Oncol. 2005; 23(23):5294–5304. [DOI] [PubMed] [Google Scholar]
- 54. Kaley TJ, Panageas KS, Mellinghoff IK, et al. Phase II trial of an AKT inhibitor (perifosine) for recurrent glioblastoma. J Neurooncol. 2019; 144(2):403–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Wen PY, Touat M, Alexander BM, et al. Buparlisib in patients with recurrent glioblastoma harboring phosphatidylinositol 3-kinase pathway activation: an open-label, multicenter, multi-arm, phase ii trial. J Clin Oncol. 2019; 37(9):741–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Wen PY, Stein A, van den Bent M, et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutant low-grade and high-grade glioma (ROAR): a multicentre, open-label, single-arm, phase 2, basket trial. Lancet Oncol. 2022; 23(1):53–64. [DOI] [PubMed] [Google Scholar]
- 57. Reardon DA, Fink KL, Mikkelsen T, et al. Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol. 2008; 26(34):5610–5617. [DOI] [PubMed] [Google Scholar]
- 58. van den Bent MJ, Brandes AA, Rampling R, et al. Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034. J Clin Oncol. 2009; 27(8):1268–1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Yung WK, Vredenburgh JJ, Cloughesy TF, et al. Safety and efficacy of erlotinib in first-relapse glioblastoma: a phase II open-label study. Neuro Oncol. 2010; 12(10):1061–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Raymond E, Brandes AA, Dittrich C, et al. Phase II study of imatinib in patients with recurrent gliomas of various histologies: a European Organisation for Research and Treatment of Cancer Brain Tumor Group Study. J Clin Oncol. 2008; 26(28):4659–4665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Reardon DA, Egorin MJ, Quinn JA, et al. Phase II study of imatinib mesylate plus hydroxyurea in adults with recurrent glioblastoma multiforme. J Clin Oncol. 2005; 23(36):9359–9368. [DOI] [PubMed] [Google Scholar]
- 62. Reardon DA, Dresemann G, Taillibert S, et al. Multicentre phase II studies evaluating imatinib plus hydroxyurea in patients with progressive glioblastoma. Br J Cancer. 2009; 101(12):1995–2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bota DA, Mason W, Kesari S, et al. Marizomib alone or in combination with bevacizumab in patients with recurrent glioblastoma: phase I/II clinical trial data. Neurooncol Adv. 2021; 3(1):vdab142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lassman AB, Wen PY, van den Bent MJ, et al. A phase II study of the efficacy and safety of oral selinexor in recurrent glioblastoma. Clin Cancer Res. 2022; 28(3):452–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Reardon DA, Brandes AA, Omuro A, et al. Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the checkmate 143 phase 3 randomized clinical trial. JAMA Oncol. 2020; 6(7):1003–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Reardon DA, Kim TM, Frenel JS, et al. Treatment with pembrolizumab in programmed death ligand 1-positive recurrent glioblastoma: results from the multicohort phase 1 KEYNOTE-028 trial. Cancer. 2021; 127(10):1620–1629. [DOI] [PubMed] [Google Scholar]
- 67. Izumoto S, Tsuboi A, Oka Y, et al. Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme. J Neurosurg. 2008; 108(5):963–971. [DOI] [PubMed] [Google Scholar]
- 68. Omuro A, Vlahovic G, Lim M, et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol. 2017; 20(5):674–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Nayak L, Molinaro AM, Peters K, et al. Randomized phase ii and biomarker study of pembrolizumab plus bevacizumab versus pembrolizumab alone for patients with recurrent glioblastoma. Clin Cancer Res. 2021; 27(4):1048–1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Zadeh G, Daras M, Cloughesy TF, et al. Ltbk-04. phase 2 multicenter study of the oncolytic adenovirus dnx-2401 (tasadenoturev) in combination with pembrolizumab for recurrent glioblastoma; captive study (Keynote-192). Neuro Oncol. 2020; 22(Suppl 2):ii237–ii237. [Google Scholar]
- 71. Desjardins A, Gromeier M, Herndon JE, et al. Recurrent glioblastoma treated with recombinant poliovirus. N Engl J Med. 2018; 379(2):150–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Cloughesy TF, Landolfi J, Hogan DJ, et al. Phase 1 trial of vocimagene amiretrorepvec and 5-fluorocytosine for recurrent high-grade glioma. Sci Transl Med. 2016; 8(341):341ra375–341ra375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Ellingson BM, Wen PY, Cloughesy TF. Evidence and context of use for contrast enhancement as a surrogate of disease burden and treatment response in malignant glioma. Neuro Oncol. 2018; 20(4):457–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007; 11(1):83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009; 27(28):4733–4740. [DOI] [PubMed] [Google Scholar]
- 76. Vredenburgh JJ, Desjardins A, HerndonJE, 2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007; 25(30):4722–4729. [DOI] [PubMed] [Google Scholar]
- 77. Awada G, Ben Salama L, De Cremer J, et al. Axitinib plus avelumab in the treatment of recurrent glioblastoma: a stratified, open-label, single-center phase 2 clinical trial (GliAvAx). J ImmunoTher Cancer. 2020; 8(2):e001146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Cloughesy TF, Brenner A, de Groot JF, et al. A randomized controlled phase III study of VB-111 combined with bevacizumab vs bevacizumab monotherapy in patients with recurrent glioblastoma (GLOBE). Neuro Oncol. 15 2020; 22(5):705–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Ellingson BM, Sampson J, Achrol AS, et al. Modified RANO, immunotherapy RANO, and standard RANO response to convection-enhanced delivery of il4r-targeted immunotoxin mdna55 in recurrent glioblastoma. Clin Cancer Res. 2021; 27(14):3916–3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Clopper CJ, Pearson ES. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika. 1934; 26(4):404–413. [Google Scholar]
- 81. Alexander BM, Ba S, Berger MS, et al. Adaptive global innovative learning environment for glioblastoma: gbm agile. Clin Cancer Res. 2018; 24(4):737–743. [DOI] [PubMed] [Google Scholar]
- 82. Ellingson BM, Gerstner ER, Lassman AB, et al. Hypothetical generalized framework for a new imaging endpoint of therapeutic activity in early phase clinical trials in brain tumors. Neuro Oncol. 2022; 24( 8): 1219– 1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Ellingson BM, Wen PY, Cloughesy TF. Therapeutic response assessment of high-grade gliomas during early-phase drug development in the era of molecular and immunotherapies. Cancer J. 2021; 27(5):395–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Ellingson BM, Harris RJ, Woodworth DC, et al. Baseline pretreatment contrast enhancing tumor volume including central necrosis is a prognostic factor in recurrent glioblastoma: evidence from single and multicenter trials. Neuro Oncol. 2017; 19(1):89–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Smedley NF, Ellingson BM, Cloughesy TF, Hsu W. Longitudinal patterns in clinical and imaging measurements predict residual survival in glioblastoma patients. Sci Rep. 2018; 8(1):14429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Ellingson BM, Bendszus M, Boxerman J, et al. Consensus recommendations for a standardized Brain Tumor Imaging Protocol in clinical trials. Neuro Oncol. 2015; 17(9):1188–1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Ellingson BM, Brown MS, Boxerman JL, et al. Radiographic read paradigms and the roles of the central imaging laboratory in neuro-oncology clinical trials. Neuro Oncol. 2021; 23(2):189–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Ahmed I, Sutton AJ, Riley RD. Assessment of publication bias, selection bias, and unavailable data in meta-analyses using individual participant data: a database survey. BMJ. 2012; 344(jan03 1):d7762–d7762. [DOI] [PubMed] [Google Scholar]
- 89. Simes RJ. Publication bias: the case for an international registry of clinical trials. J Clin Oncol. 1986; 4(10):1529–1541. [DOI] [PubMed] [Google Scholar]
- 90. Sterne JA, Egger M, Smith GD. Investigating and dealing with publication and other biases in meta-analysis. BMJ. 2001; 323(7304):101–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Sutton AJ, Duval SJ, Tweedie R, Abrams KR, Jones DR. Empirical assessment of effect of publication bias on meta-analyses. BMJ. 2000; 320(7249):1574–1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Song F, Parekh S, Hooper L, et al. Dissemination and publication of research findings: an updated review of related biases. Health Technol Assess. 2010; 14(8):1–220. [DOI] [PubMed] [Google Scholar]
- 93. Lukas RV, Rodon J, Becker K, et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J Neurooncol. 2018; 140(2):317–328. [DOI] [PubMed] [Google Scholar]
- 94. Batchelor TT, Duda DG, di Tomaso E, et al. Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol. 2010; 28(17):2817–2823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Brown N, McBain C, Nash S, et al. Multi-center randomized phase ii study comparing cediranib plus gefitinib with cediranib plus placebo in subjects with recurrent/progressive glioblastoma. PLoS One. 2016; 11(5):e0156369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Wen PY, Drappatz J, de Groot J, et al. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients naive to antiangiogenic therapy. Neuro Oncol. 2018; 20(2):249–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



