Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2024 Aug 1.
Published in final edited form as: Nat Rev Cancer. 2024 Jun 19;24(8):578–589. doi: 10.1038/s41568-024-00708-4

Roadmap: Why do patients with cancer die?

Adrienne Boire 1,ǂ, Katy Burke 2,ǂ, Thomas Cox 3,*,®, Theresa Guise 4,ǂ, Mariam Jamal-Hanjani 5,6,7,ǂ, Tobias Janowitz 8,ǂ, Rosandra Kaplan 9,ǂ, Rebecca Lee 10,11,ǂ, Charles Swanton 6,7,12,ǂ, Matthew G Vander Heiden 1,14,ǂ, Erik Sahai 10,*,®
PMCID: PMC7616303  EMSID: EMS197473  PMID: 38898221

Abstract

Cancer is a major cause of global mortality, both in affluent countries and increasingly in developing nations. A large number of patients with cancer experience reduced life expectancy and have metastatic disease at time of death. However, the more precise causes of mortality and patient deterioration prior to death remain poorly understood. This scarcity of information, in particular the lack of mechanistic insights, presents a challenge for the development of novel treatment strategies to improve the quality of , and potentially extend life, for patients with late-stage cancer. In addition, earlier deployment of existing strategies to prolong quality of life is highly desirable. In this Roadmap, we review the proximal causes of mortality in patients with cancer, discuss current knowledge about the inter-connections between mechanisms that contribute to mortality, before finally proposing new and improved avenues for data collection, research, and the development of treatment strategies that may improve patients’ quality and duration of life.

Introduction

The phrase “metastasis accounts for 90% of cancer deaths” is one of the most widely used in cancer research, yet it is overly simplistic, imprecise, and it is difficult to find any primary analysis supporting the statement. Whilst patients with metastatic disease are overwhelmingly more likely to die than patients with non-metastatic cancer(1,2), the determinants of cancer mortality are multifaceted and frequently involve dysfunction of multiple interconnected systems within the body. Understanding the mechanisms underpinning the causes of mortality, and subsequently intervening, has the potential to make cancer a less destructive disease, improving both the quality and length of life for patients with cancer. However, systematic analyses of the acute and root causes of mortality in patients with cancer are scarce, in part because death certificates rarely record enough information to understand the exact reason why the patient died beyond them having a malignancy. Potentially concomitant comorbidities are also not fully recorded, including in most cases the precise event that led to death. Instead, causes of death may be simply listed as “metastatic carcinoma” or “complications of cancer” which give little insight into why a patient actually died. Even in cases where the cause of death may be attributed to a single event, for example a thromboembolism, the underlying cause of that specific event may be complex. Indeed, metastatic cancer leads to perturbed function of multiple organ systems, and importantly, not just the organs to which disease has spread. This is likely due to the exuberant activation of local and systemic inflammatory, tissue repair, and immune-suppressive programmes.

A simple view would be that the death from metastatic disease correlates with the burden of disease. However, evidence suggests that the situation is more complex, with many factors influencing how metastases impact vital functions and ultimately lead to death. Firstly, metastases to different organs will lead to different impacts on overall health. For example, brain metastases can lead to dysfunction of the central nervous system, whereas peritoneal metastases may cause obstruction of the bowel. In addition, the size or extent of metastases may not necessarily correlate with dysfunction of the organ where it is located (3). Second, the production of the molecular mediators of organ dysfunction can vary between metastases and cancers of different origins. Third, individual patient characteristics like age, sex, overall health, pre-existing comorbidities, genetics and socio-economic status vary (4). Together, these factors directly influence the course of, and physiological response to metastatic disease, and can have profound indirect effects by limiting available treatment options and/or the ability of patients to tolerate or complete all intended treatment (5,6). To understand why patients with cancer die, a closer examination of the factors contributing to mortality in patients with, and dissection of the intricate web of causes that shape the frequency and dynamics of death are required. In this Roadmap, we briefly review data considering the immediate causes of mortality, highlight the intricate inter-connections between different aspects of patient deterioration, and conclude with recommendations for future studies of late-stage cancer that may shed new light on this important aspect of cancer biology and medicine. Death may be related to an acute event, but the underlying mechanisms which trigger it may be modifiable or even preventable. In addition, other deaths may be the end stage of a continuum of deterioration, allowing the possibility of targeted intervention to improve quality of life. In addition, it has been noted that early palliative care improves survival (7). Ultimately, increased understanding of the processes occurring in patients with advanced disease should lead to improved strategies to minimise ill-health and suffering at the end of life. Coupled to this, patients and those around them should be enabled to have essential discussions about their wishes and preferences, minimising potentially inappropriate treatments and maximising quality of life (8).

Acute events leading to mortality

Although some cancers can be considered a chronic disease, with many patients living with their disease for years, the immediate cause of mortality can often be an acute event. Here we briefly summarise common acute events leading to death in patients with cancer (Figure 1). Whilst it is not possible to precisely determine, it is likely that the acute causes discussed below may account for up to half of cancer deaths (9,10). Immediate causes of mortality in other patients are less clear, with a more gradual deterioration typically occurring in vital organ systems.

Figure 1. Illustration of the proximal causes of mortality in patients with cancer.

Figure 1

Image shows organs that frequently become dysfunctional in late stage cancer patients.

Vascular/coagulation/cardiac failure

Patients with cancer are at an elevated risk of thrombo-embolism [G], which may trigger respiratory failure, fatal strokes, heart failure or myocardial infarction (11). In some cases, disseminated intravascular coagulation [G] can lead to thrombotic obstruction of small and midsize vessels leading to organ failure (12). Haemorrhagic complications from depletion of platelets, via either immune or non-immune mechanisms PMID: 19466980 PMID: 31205603, and reduced levels of coagulation proteins can also be life-threatening (12). Congestive heart failure [G] can also be a proximal cause of mortality, although the underlying causes are complex and include loss of cardiac muscle (associated with cachexia), shifts in intravascular fluid status, and thrombo-embolic events (13). Interestingly, bone metastases are particularly associated with cardiovascular problems, although the underlying mechanism remains unclear (14). Comorbidities affecting the cardiovascular system may make patients more prone to such events. Spatial occlusion or invasion into vessels by cancer metastases can also lead to failure in blood supply or catastrophic haemorrhage (1518).

Displacement, functional impairment or obstruction of vital organs

The volume of disease may impair the function of a vital organ. This can be the case with brain metastases and glioblastoma or other primary brain cancers, with either extensive invasion, brain herniation [G], or oedema resulting in midline shift [G] or increased intracranial pressure irreversibly compromising brain function (1820). In addition, patients may develop seizures, which if uncontrolled, can result in death (21,22). However, this does not apply to all brain metastases, with leptomeningeal metastases having minimal impact on intracranial pressure and brain structure; instead, these commonly obstruct cerebrospinal fluid flow and/or affect nerve function resulting in hydrocephalus, deterioration of neurological function, and death (22).

Large lung metastases may impair the essential function of gas exchange. However, patients with miliary-like disease – characterised by nodules too numerous to count – can live with extensive disease in an organ with surprisingly little impact on function until a hard-to-predict tipping point is reached, which is then followed by rapid deterioration (23). As with brain metastases, the volume of disease is often not sufficient to account for organ failure, as even relatively small volume (<100ml lung metastases, compared with 4-5l total lung volume) can be fatal (24). Lung oedema [G] related to other pathology such as infection or heart failure can also impair gas exchange causing death, and pleural effusions are an additional common contributor to death. Pleural effusion may be related to presence of disease within the pleura as opposed to total tumour volume (25,26).

Bowel obstruction can be a cause of mortality, particularly in patients with peritoneal disease as found in ovarian, colorectal and gastrointestinal cancers (27). Both liver and kidney failure will also cause death in patients with cancer. Reasons for the failure of these organs include obstruction of the bile duct or ureters by metastases, therapy-induced toxicity leading to compromised normal organ function (discussed below), and reduced tissue perfusion due to hypotension or dehydration (2831). In addition, sepsis can result from obstruction of the bile ducts or ureters, which occurs unpredictably and often progresses rapidly leading to multiple organ failure and ultimately death.

Infections

Bacterial infections are the most common infection in patients with cancer, due to impaired immune systems resulting from both the cancer itself as well as certain cancer treatments (discussed in detail in the Iatrogenic effects section), which induce myelosuppression and leukopenia. Patients with cancer can have an elevated risk of opportunistic viral, fungal and protozoal infections, which would typically be considered mild in healthy individuals, but which can cause serious life-threatening complications in those with cancer. Pneumonia and other lung infections leading to respiratory failure are often listed as causes of mortality in patients with cancer (32,33). One of the most striking recent examples of this is the increased mortality observed in patients with cancer, particularly haematological cancers, who succumbed to COVID-19 more than the general population (33,34).

Paraneoplastic syndromes

Paraneoplastic syndromes are a group of rare disorders that can occasionally cause irreversible damage to critical organs and death. They are most associated with lung, breast, ovarian, and lymphatic cancers, causing tissue or organ dysfunction at sites distinct from the location of the tumour. A variety of mechanisms underpin paraneoplastic syndromes, including the inappropriate production of cytokines, hormones, and antibodies. For example, excess PTHRP production by tumours can lead to hypercalcemia [G]. Inappropriate anti-diuretic hormone production is commonly associated with small cell lung cancer resulting in hyponatraemia and some neuroendocrine pancreatic tumours (insulinomas) secrete large amounts of insulin (3538). Tumours can also trigger the aberrant production of autoantibodies leading to Lambert-Eaton Myasthenic Syndrome, N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis [G], and Myasthenia Gravis (39). Whilst treatment can usually manage the symptoms, however in a subset of cases the syndromes cannot be controlled and are fatal (40).

Therapy-induced toxicity

Although therapies are developed and administered with the intent of primarily targeting the tumour, almost all have some detrimental impact on normal tissue function. In some cases, the unintended consequences of therapy can be life-threatening. Auto-immune reactions resulting from targeting immune checkpoints can have fatal consequences, including myocarditis and encephalitis (4143). Death can result from acute neutropenic sepsis related to chemotherapy (44). Depletion of platelets as a result of therapy can lead to fatal bleeding (45). Arrhythmias, cardiomyopathy and coronary vasospasm [G] are also a cause of death related to some anti-cancer treatments such as 5-flurouracil and capecitabine (4648). The long-term detrimental effects of some therapies are discussed in detail in the section on Iatrogenic effects.

Underlying causes

Determination of the proximal cause of mortality prompts further questions around the underlying factors giving rise to lethal pathology, and ultimately how metastatic cancer triggers or accelerates those factors. In this section, we consider how chronic disruption of three major physiological/organ systems are perturbed in patients with cancer and how these might contribute to mortality.

The immune and haematopoietic system

In patients with cancer, the immune system becomes progressively less able to mount effective responses to infectious challenge, a phenomenon often generically termed "immune exhaustion" (this usage is distinct from the more specific usage of immune exhaustion as a failure of tumour-reactive T-cells to function). As a result, patients with metastatic disease have increased susceptibility to a wide range of infections, and typically suffer more severe consequences than would otherwise be observed in healthy individuals (49). Multiple mechanisms contribute to the reduced capability of the immune system to respond to infection. The presence of cancer cells in diverse organs triggers similar cellular and molecular events to wound responses (50). The production of cytokines including IL6, G-CSF, and GM-CSF, both by tumour cells and other tumour microenvironment (TME) cells, perturbs haematopoiesis leading to altered profiles of leukocytes(51). While in the short term, this may have limited consequences on the body’s ability to respond to other challenges, prolonged disruption to haematopoiesis can strain the ability of haematopoietic stem cells (HSCs) to generate sufficient cells of the right type to cope with infections, with increased myeloid to lymphoid cell ratios. Clonal haematopoiesis [G] can be increased in patients with cancer, with myeloid skewing of immune cells and overall myeloid mediated immune suppression and diminished naïve T cell reservoirs (51). Reduced production of platelets and altered iron metabolism leading to compromised oxygen carrying by red blood cells is also observed in many patients (52). Other problems, such as immunoparesis [G] can arise, with a high frequency observed in multiple myeloma patients (53). Once again, comorbidities leading to either immune suppression or auto-immunity can intersect with the detrimental effects of cancer on the immune system. T-cell responses to infection are impaired in the presence of cancer with decreased proliferation and expression of granzyme B typically observed (54). The chronic stimulation of T-cells with neoantigens arising from ongoing mutational processes may also contribute to their weakened functionality. Moreover, immune surveillance of tumours inevitably selects for the production of immune suppressive factors by cancer cells that further compound the issue (55).

Other consequences of cancer result can indirectly result in increased likelihood of infection. For example, vessel obstruction from cancer results in decreased flow of fluids such as bile, urine and lymph, creating environments in which bacteria can thrive (56). Blockage of the bronchial tree can lead to pneumonia (57). The invasive phenotype of cancer can result in fistula [G] formation (e.g. recto-vaginal in colorectal cancer) which enables bacteria to invade (58) from one body cavity to the next facilitating spread and subsequent systemic spread leading to sepsis. Furthermore, patients are often rendered bedbound or have limited mobility as cancer progresses, resulting in increased chance of infections through decreased respiratory ventilation and atelectasis [G], as well as pressure sores and oedema (59).

Disruption to haematopoiesis can also contribute to defects in coagulation and haemostasis. Elevated platelet numbers, termed thrombocytosis is found in cancer patients and correlated with higher mortality. The altered inflammatory cytokine milieu caused by the tumour may promote megakaryopoiesis, potentially through increasing Thrombopoietin (TPO) production by the liver, and leading to higher platelet numbers. The risk of clotting can be further increased by the production of tissue factor [G], which is responsible for initiating the clotting cascade, by tumour cells (60). These mechanisms increase the likelihood of fatal thromboembolisms (60).

Iatrogenic effects [G] also play a role in the reduced immune function in patients with cancer. Cytotoxic therapies interfere with the proliferation and division of haematopoietic stem cells and can leave the immune system unable to mount effective responses to pathogens, leading to mortality (61). In severe cases, pancytopenia results, marked by a significant decrease in all three major blood cell lineages (red cells, white cells and platelets) (62). This can lead to severe anaemia, increased infection susceptibility, and increased likelihood of bleeding (44,63,64). In other cases, more limited subsets of haematopoietic cells are affected. Thrombocytopenia – low platelet levels – leads to hypo-coagulation and elevates the likelihood of haemorrhage (63). Thus, during cancer development and treatment, haemostasis mechanisms may be either augmented or attenuated, and in both cases the end result is less predictable and well-controlled coagulation. Neutropenia – low neutrophil levels – renders patients less able to fight infection and contributes to cancer mortality from infections that in many cases are thought to arise from resident mucosal flora (65). Treatments, including chemotherapy and radiotherapy, often result in the breakdown of mucosal barriers (e.g. oral mucositis) resulting in higher numbers of infections from pathogens which normally reside on these surfaces (66). In addition, corticosteroids, which are often given to alleviate symptoms or manage toxicity, can also add to suppression of immune response and compound the risk of infections in patients (67). Clonal haematopoiesis, which is already more frequent in cancer patients, can be further increased by chemotherapy (68). More generally, cancer therapies can increase aging-associated processes and reduce organ function (69). The wide-spread use of corticosteroids, used to counteract some of the side-effects of therapy and to reduce the symptoms of cancer, further suppresses the immune system. Opioid pain relief administered to those with late-stage disease can also suppress the function of various bodily systems (70). Finally, infections can arise due to the insertion of drains and stents, or central venous catheters (CVC, also known as lines) for delivery of therapies. Infections from lines is estimated to be around 0.5–10 per 1,000 CVC-days (71,72).

Immunotherapies present a different set of immune complications from conventional therapies. These primarily relate to over-activation of the immune system leading to auto-immunity and, in some cases, cytokine storms that are treated with anti-cytokine therapies such as tocilizumab, anakinra and ruxilitinib, all of which can further suppress the immune response (73). However, deaths attributable to autoimmune side effects of checkpoint inhibitors are rare (approximately 1%) especially if toxicity is managed promptly (74,75). Colitis is a frequent problem, with disruption to colonic barrier function leading to increased susceptibility to perforation, which can be life threatening. In addition, Guillain-Barré syndrome [G], hepatitis, and myocarditis are also causes of checkpoint inhibitor-related deaths (7678). Once again, high dose corticosteroids are the main first line treatment to manage autoimmune side-effects in patients receiving immunotherapy. A subset of patients experience hyperprogressive disease [G] following immunotherapy, the reasons for this are still being delineated but there is likely a role for innate lymphoid cells releasing pro-growth cytokines (79). Cell-based immunotherapies can also lead to disrupted bone marrow function and subsequent myelosuppression (80).

The nervous system

The brain serves as a central nexus, orchestrating all vital functions. It is the hub of thought processes, emotions, and sensory perception, and regulates, directly or indirectly, everything from heartbeat and breathing, to appetite. In addition to physical disruption of brain structure and intracranial pressure (discussed in the section on immediate causes of mortality) (81), brain metastases impact the nervous system in multiple ways. Tumours in the brain or its surrounding tissues can significantly impair neural connections, leading to cognitive deficits, motor/sensory dysfunction, and even personality changes (8183). Interactions between brain metastases and neurons lead to changes in cortical function (8486). Even in regions of the brain without overt metastases, neuro-excitability can be increased, leading to changes in cognition, alertness, and mood (87). Tumours can slow the posterior dominant rhythm, leading to reduced alertness, loss of working memory and deterioration of quality of life (88). Circadian rhythms are also impacted, leading to problems in memory and sleep, which is vital for the body’s repair processes that are essential for overall health and functioning (89). Ultimately, many of these changes are not sustainable long-term. How these changes may lead to death is unclear, but it may follow similar trajectories to those in dementia patients.

Brain function can also be disrupted in patients without brain metastases, with autonomic nervous system dysfunction often reported (90). Intriguingly, anhedonia – a lack of ability to experience pleasure – occurs in many patients (91). The mechanistic causes of this are unclear, but it is not restricted to patients with brain metastasis suggesting that circulating systemic factors may play a role. The wider effects of metastatic cancer on patient’s mental wellbeing are discussed in Text Box 1. However, beyond an effect on well-being, the disruption of brain function can contribute to anorexia, and reduced nutrition can influence many other physiological and pathophysiological processes (92,93).

TEXT BOX 1. Psychosocial and societal factors contributing to the deterioration of late-stage cancer patients.

Psychological and social factors can have major and wide-ranging impacts on patients with incurable cancer. This is manifest in over three-fold higher suicide rates in cancer patients, with even higher rates in women (97). Of note, these rates were further exacerbated in less advantaged sociodemographic groups, arguing that financial and possibly healthcare access problems are linked to suicide in cancer patients. However, psychological symptoms in cancer patients are far more extensive than those captured in studies of suicide. Anhedonia and depression are frequent in cancer patients, impacting their overall well-being, treatment adherence, and outcomes including mortality (98). These psychological challenges often intertwine with physical symptoms, compounding the burden of each (99). Several studies have linked stress-related psychosocial factors to cancer mortality (100), with recent work beginning to uncover the cellular and molecular mechanisms at play (101).

Research on the psychosocial aspects of cancer care, including emotional and cognitive well-being, remains under-emphasized. Barriers to the integration of psychosocial care into cancer care include stigma, difficulty identifying significant distress, limited access to evidence-based psychosocial treatments and concerns about cost (102) . An integrated system of psychosocial care including population-based screening and targeted treatment and access to good-quality palliative care improves emotional wellbeing (103) and physical symptoms (104) and is likely to be cost-saving (105). A deeper understanding of the mechanisms underlying neuropsychological systems and insights into how metastatic disease impacts the physiochemical axes will be crucial. Such insights could inform tailored interventions, therapies, and support structures that address the emotional toll of cancer, enhancing the holistic care approach, and improving quality of life. Expanding psychosocial research can help bridge gaps in addressing mental health in cancer, ultimately improving patients’ quality of life during and after treatment (106,107).

The role of the peripheral nervous system [G] in cancer-related death is not well described. While the burgeoning field of cancer neuroscience provides evidence that the efferent system can support local and metastatic tumor growth (9496), at this time, it is unclear if the reverse is also true. There is clear evidence of autonomic nervous system dysfunction in patients with cancer (90), raising the possibility that cancer-mediated interruption of afferent impulses might impact overall survival. Further studies are needed to explore this possibility.

Metabolism and cachexia – catabolic effects of cancer

The presence of metastases presents altered energetic and anabolic demands on the body, leading to detrimental imbalances in metabolism (108). Progressive and involuntary loss of body weight – termed cachexia – is a widespread multiorgan phenomenon commonly seen in patients with metastatic cancer (108110). This complex syndrome is characterized by a net negative energy balance, driven by the combination of increased energy expenditure and catabolism, with reduced appetite and caloric intake. Persistent decrease in nutrient intake is a key component across patients with many different cancers, leading to breakdown of host tissues, with loss of adipose tissue and muscle mass varying between patients and among different cancers. However, the contribution of increased energy expenditure (as a result of tumour burden) is less clear. Sarcopenia [G] may be particularly prominent in some patients, possibly representing an independent pathology from other more global tissue wasting phenotypes, and in extreme cases, loss of cardiac or intercostal muscle mass can be fatal due to insufficient cardiac and/or respiratory function (111,112). These events have also been observed in the context of extreme starvation in patients with non-cancer conditions; for example, anorexia nervosa, where cardiac dysfunction, in particular fatal bradycardia and sinus pauses, can cause pulseless electrical activity and death (113,114). Electrolyte disturbances and hypoglycaemia that are often observed in cases of severe malnutrition may exacerbate the risk of such arrhythmias (113). Cachexia also has effects on other organs, including the brain and immune system. Compromised immune function is a major consequence of starvation-induced tissue wasting, and suggests that altered systemic metabolism leading to, or associated with cachexia, may be a contributor to the immune dysfunction present in some patients with cancer (115). Conversely, several studies have shown that both the brain and immune system can contribute to cachexia (109,110).

Cachexia is multifactorial and has many potential causes. In some limited cases, tumour metabolism leads to systemic changes that increase energy usage. For example, high levels of lactate secretion by tumours can trigger the liver to convert lactate back to glucose, which requires energy input – termed the Cori cycle (116). Such cycles can increase metabolic demand on the liver leading to further perturbation of liver function. However, cachexia does not correlate with disease volume in many cancer types (117). Thus, it is hard to reconcile a model in which the energetic and catabolic demands of the volume of disease are the main trigger for cachexia. Numerous studies have begun to reveal the possible molecular underpinnings of cachexia in some cancer types. Disruption of signalling by TGFβ and related ligands is a recurring theme (118120). For example, circulating GDF15, a highly conserved member of the TGFβ family, is a known mediator of anorexia and weight loss, and increased circulating levels in patients with lung cancer have been shown to correlate with cachexia development (121). TGFβ itself can also promote muscle loss via the induction of myostatin (122). Induction of signalling by activin – another TGFβ-family ligand – can also have similar effects on muscle mass (123,124). Furthermore, modulation of RyR1 downstream of TGFβ can perturb sarcomere organisation and thereby lead to muscle weakness (125). As such, pre-clinical studies have demonstrated the potential utility of TGFβ blockade in preventing cachexia (126).

Elevated levels of cytokines, including TNFα, IL1, and IL6, can also play roles in cachexia (127129). TNFα induces multiple aspects of cachexia (130). Muscle wasting is promoted through increased TNFα and NFκB-dependent ubiquitin-mediated proteolysis of muscle protein (131,132). IL6 triggers muscle loss through a similar mechanism. Lipid metabolism is impacted by TNFα reducing the expression of lipoprotein lipase and free fatty acid transporters, thereby reducing the accumulation of fat (133). TNFα can also reduce appetite through the production of corticotropin-releasing hormone [G] (CRH). IL1, which triggers similar proximal changes in cell signalling to TNFα, can activate many of the same processes (133). It is also interesting to note that TGFβ, IL1, and IL6 are associated with programmes in cancer cells that drive metastasis, which could potentially explain why metastatic disease is linked to cachexia more strongly than the presence of primary disease alone.

Whole body dysfunction

Although consideration of different organ systems is useful for highlighting some of the key events contributing to cancer mortality, the inter-connected nature of body systems and the pleiotropic characteristics of the molecular mediators at play mean that ultimately it is essential to consider whole body dysfunction when thinking about causes of cancer mortality. Furthermore, such analyses may explain cancer deaths without an acute proximal cause. As discussed above, cytokines with potent effects on the immune system, as well as effects on appetite, can be contributors to cachexia. Therefore, it is unsurprising that tumours impact both immune and metabolic function. The immune and nervous systems are highly sensitive to metabolite availability; for example, the brain has a high demand for glucose (115,134). Several factors, including lactic acid production and kidney dysfunction can lead to life-threatening systemic acidosis in patients with cancer, particularly haematological malignancies with high cell turnover (135). These can be further exacerbated upon initiation of cytotoxic therapy resulting in tumour lysis syndrome which can be fatal (136). Consequently, metabolic perturbations and cachexia impact these systems. Over time, the cumulative stress of metabolic alterations caused by metastases, chronic changes in the level of cytokines, constant generation of tumour (neo)-antigens, aggressive therapies, and incidental infections lead to exhaustion of the adaptive immune system and hamper the regenerative capacity of many organ systems with debilitating effects (14). This multi-faceted burden can ultimately trigger a body-wide shut-down leading to death.

Are cancer mortality causes cancer-specific?

Although a subset of mortality causes are cancer-specific, such as metastatic invasion compromising specific organ function, the progressive and inter-connected deterioration of multiple organ systems likely underlies many cancer deaths. This may be further influenced by interaction with other co-morbidities. Of note, similar progressive deterioration is sometimes observed in the context of chronic infection and inflammation, with both cachexia and immune exhaustion being associated with diseases such as tuberculosis (TB) and Human Immunodeficiency Virus (HIV) infection (137139). This raises the question of whether the causes of death in patients with cancer are specific to cancer, or whether cancer (or any other chronic disease) is simply an accelerant of aging processes occurring in healthy individuals. This hypothesis has practical implications because, if proven, it would suggest that lessons and approaches from other disease contexts could be readily transferable to patients with metastatic cancer. For example, the targeting or modulation of senescent cells is an active area of anti-aging research and numerous pre-clinical studies have indicated that similar strategies can attenuate the systemic effects of cancer (140142).

Recommendations

The goal of this Roadmap is to propose ways to improve our understanding of why patients with cancer die and thereby develop better strategies to ameliorate symptoms and prolong life with good quality in cancer patients. To this end, we propose that the following steps would be useful.

Improved records and reporting

It is notable how infrequent systematic reviews of the precise causes of cancer mortality are. This gap in knowledge, and recognition that this is often simply not known, is a major hindrance to learning and progress. Although improved accuracy of reporting on death certificates would be desirable, it would require a shift in longstanding clinical habits, and may not be easily achievable in healthcare systems under strain. Palliative care primarily focusses on symptom control for patients whilst balancing the potential benefits and burdens of additional diagnosis. Nevertheless, to address the gaps in our knowledge, it would be desirable to fund and establish prospective studies that continue active monitoring of patients as they transition from active disease treatment to palliative care. If possible, monitoring should be non-invasive to not compromise patient comfort at the end of life. The great advances being made in patient monitoring with wearable technologies [G] might facilitate this, and could be used for earlier detection of infections enabling quicker intervention. Caregiver involvement in reporting of symptoms may also play a role. Patient/public involvement in this type of research will be critical. In addition, consent to obtain more detailed information from the community/palliative care teams on the contributing factors to death would provide further insight. In addition to information gathered prior to death, research autopsies have the potential to shed further light on the aetiology of death, such as thromboembolic events that may not have been detected in the absence of symptoms or diagnostic testing – discussed in Box 2. Furthermore, the availability of post-mortem samples can aid research into the biological underpinnings of metastases and processes leading to death. The greatest amount of information would be gained from cohorts additionally enrolled into warm autopsy programmes (see Text Box).

TEXT BOX 2. Research Autopsy Programmes and their optimisation.

Research autopsies are initiatives that involve the prompt collection of tissues from deceased individuals shortly after death, while tissue morphology is intact, and cells and tissues have not undergone significant post-mortem changes. Research autopsy studies can be labour intensive, and care is required in their logistical planning. The post-mortem interval (PMI) to autopsy can vary depending on the infrastructure available and can have implications for the utility of samples collected after death. For example, shorter PMIs achieved in rapid warm autopsy studies can more effectively facilitate in vitro (e.g. cell line) and in vivo (e.g. organoid and xenograft) models, and can derive better quality RNA (143,144). However, such studies are not easily established in the absence of out-of-hours facilities and expert input. Autopsies performed with longer PMIs, for example up to several days after death, have been shown to have maintained tissue morphology and adequate DNA and RNA to facilitate cellular imaging techniques and genomic sequencing approaches (145,146). Therefore, there is merit and general scientific value with autopsies regardless of the PMI, provided consideration is given to the question being addressed, and the experimental approach.

The most powerful data are obtained from patients already involved in clinical studies prior to death. Information about disease course, longitudinal scans, tissue and blood analysis (cell counts, electrolytes, cytokines, metabolites, and possibly ctDNA) greatly enhances what can be learnt from post-mortem tissues. Sensitivity is required to align the desire to acquire data with the wishes of the patients and their families, such that ultimately each autopsy has the potential to be meaningful and shed light on the biological processes leading to death.

More detailed observational clinical studies

Disease burden is not well correlated with survival; however, we propose that the accurate identification of prognostic factors correlating with survival should provide important insights into what ultimately precipitates mortality. As the cost of both targeted and non-targeted analysis of proteins and metabolites decreases, it should also become more feasible to explore molecular predictors of survival. Once identified, such factors could then be monitored in a targeted way prospectively with the potential to intervene upon where possible. In this setting, both the tumour and patient trajectory would receive precision tailored treatments, the impact of which would need to be studied in randomised controlled trials. Even in the context of early phase trials, additional data could be obtained about patient symptoms in addition to safety considerations and tumour burden. Clinical imaging could also be exploited. Many patients receive CT and PET scans and these contain abundant information about the burden and location of metastases and offer the opportunity to study changes in extent of adipose and muscle tissue and therefore body composition in relation to cachexia. Machine learning and artificial intelligence can be capitalised on to accurately measure these parameters, meaning that what would have previously been prohibitive due to the hours of radiologist time required is now feasible (147,148). In addition to the analysis of scans, the application of machine learning approaches to metabolite, cytokine, immune cell, and wearable technology-derived multi-modal, and multidimensional data may also uncover previously unknown parameters that correlate with mortality (149). As outlined in Text Box 1, incorporating psychosocial metrics into the study of late-stage cancer could also enable improvements in patients’ mental well-being.

Increasing the relevance of model systems

Pre-clinical models will also have a place in determining the linkage between events found to precede death and cause of death; however, there should be an emphasis on reverse translation of questions from human studies to pre-clinical models. By way of example, this could involve modelling how metastases impinge on the body’s ability to respond to infection by challenging metastatic models with a pathogen. Animal ethics and husbandry considerations mean that mice are housed in controlled environments where exposure to pathogens is rare, and the types of pathogen exposure very narrow, so this type of information is currently lacking. To be optimally informative, practical and ethical complications around studying end-of-life physiology seen in patients need to be considered. Most models are chosen for their rapid progression, often with less than a month between primary or metastatic tumour seeding and death. These are not optimal for studying longer timescale chronic changes in patients. The development of slower progressing models, implementation of multiple lines of treatment and mimicking presence of other co-morbidities should enable models to more accurately recapitulate observations made in patients. Furthermore, most pre-clinical cancer research currently uses young mice that fail to accurately mirror the interplay between aging and cancer seen in humans. Researchers need to recognise the importance of and adopt more age-appropriate mouse models to better understand cancer mortality. In addition, most studies focus solely on tumour burden (which may only be possible at the point of death rather than dynamically) or tumour size as a marker of disease due to the technical challenges of accurately quantifying organ impairment. Furthermore, when survival is reported in mouse studies it is often animal care facility driven ethically humane end-points that mandate euthanasia as the cause of death. Tumour volume response and progression are poor surrogates of mortality in patients (150), therefore better modelling of other metrics of tumour activity and impact on the body system may lead to better drug devlopment. While minimizing and alleviating suffering in experimental animals is critical, ethical considerations limit the ability to study mortality in mice. Thus, an expanded repertoire of analysis would help to understand how metastases impact specific systems and events, including the haematopoietic and nervous systems, as well as whole-body physiology and metabolism. Analysis of small volumes of blood can provide data on metabolites and cytokines, as well as complete blood counts (red blood cells/white blood cells/ Ppatelets) while increasingly sophisticated and automated technology is available to monitor mouse behaviour. It is worth noting that weight loss is frequently used as a humane end point, which indicates that many cancer models trigger cachexia and that with appropriate measurements there is an opportunity to learn more about this phenomenon in existing models. We advocate more detailed reporting of why mice were culled in experimental studies – e.g. tumour volume, weight loss, laboured breathing, complete blood cell counts and blood chemistry.

Clinical trials

The types of analyses detailed above will provide correlation between different factors and mortality, but not causative linkage. Ultimately, this information depends on testing in the context of clinical trials. Many of the mediators of immune dysfunction and cachexia can now be targeted with function blocking antibodies or forms of receptor traps, and are being actively explored in clinical trials. Several of these interventions were originally developed for chronic inflammatory conditions, which further highlights links between cancer and inflammation. The use of appropriately chosen secondary end-points would provide an opportunity for testing whether correlative associations have a causal basis. In addition, many cancer drug trials stop providing an intervention at the point where a cancer progresses. The mechanisms behind cancer cachexia suggest that trials should be adapted to additionally consider clinical benefit in terms of weight/muscle loss/other specific determinants of efficacy, rather than to solely monitor cancer progression.

Concluding remarks

While efforts at cancer prevention and the development of curative treatment rightly receive considerable attention, we argue that understanding the precise events leading to cancer mortality should not be overlooked by funding bodies. Understanding the causes of dysfunction across multiple organ systems, may provide novel strategies to manage symptoms of advanced cancer. In addition, better knowledge of the processes leading to death could enable patients and those around them to have essential discussions about their wishes and preferences, minimising potentially inappropriate treatments and maximising quality and enjoyment of life. Further, more precise biomarkers of the likely timing of death may enable patients and their families to better utilise the time that is left. In the longer term, strategies to prevent organ dysfunction should offer considerable benefit to both patients with high tumour burden and those who have low disease burden but die from factors produced by cancer.

Glossary

Atelectesia

partial collapse or incomplete inflation of the lung

Brain Herniation

Pressure-induced movement of brain tissue

Clonal Haematopoiesis

An aging-associated process in which haematopoiesis becomes dominated by one or a small number of genetically distinct of stem or progenitor cells. Clonal haematopoiesis is linked to an increased risk of haematological malignancies

Congestive Heart Failure

Inability of the heart to pump blood properly

Coronary vasospasm

Constriction of the arteries supply blood to the heart

Corticotropin-Releasing Hormone (CRH)

One of the major factors that drives the body’s response to stress

Disseminated intravascular coagulation (DIC)

DIC is a rare but serious condition where abnormal blood clotting occurs throughout the body’s blood vessels

Encephalitis

Inflammation of the brain

Fistula

An abnormal connection that forms between two body parts, such as an organ or blood vessel and another often unrelated structure in close proximity

Guillain-Barré syndrome

This syndrome is a rare disorder in which your body’s immune system attacks your nerves that can lead to paralysis

Hypercalcemia

Elevated calcium levels in the blood, often caused by overactive parathyroid glands. Hypercalcemia is linked to kidney stones, weakened bones, altered digestion, and potentially altered cardiac and brain function

Hyperprogressive Disease (HPD)

Rapid tumour progression sometimes observed during immune checkpoint inhibitor (ICI) treatment

Iatrogenic effects

Harm caused by cancer treatments, often unavoidable

Immunoparesis

Defined as the marked suppression of polyclonal immunoglobulins in the body

Lung Oedema

Lung or pulmonary oedema is a condition caused by excess fluid in the lungs. This fluid collects in the alveoli compromising function and making it difficult to breathe

Midline Shift

The observation of displacement of brain tissue across the centre line of the brain, suggestive of uneven intracranial pressure

Paraneoplastic syndromes

A group of rare disorders that occur when the immune system reacts to changes in the body triggered by the presence of a neoplasm

Peripheral Nervous System

A dense network of nerves that transmit information from the brain (efferent neurons) to the periphery and conversely transmit information from the periphery to the brain (afferent neurons)

Sarcopenia

Sarcopenia is a condition characterised by loss of skeletal muscle mass and function

Thrombo-embolism

The lodging of a circulating blood clot within a vessel leading to obstruction. Thrombo-embolisms may occur in veins (venous thrombo-embolism) and arteries (arterial thrombo-embolism)

Tissue Factor

a key component of the pathway regulating blood clotting, specifically the receptor and cofactor for factorVII/VIIa

Wearable technologies

Devices worn on the body, typically in the form of accessories or clothing, that incorporate advanced electronics and technology to monitor, track, or enhance various aspects of human life. Examples include smartwatches and fitness trackers

Figure 2. Recommendations for improving understanding of causes of cancer mortality.

Figure 2

Scheme shows how recommendations can interlink to provide both improved understanding of the underlying biology and strategies to improve patient’s quality of life.

Funding and Disclosures

A. Boire is funded by National Institutes of Health/National Cancer Institutes Cancer Center Support Grant P30 CA008748.

A. Boire reports no conflicts of interest.

K. Burke is employed by the UK National Health Service.

K. Burke reports no conflicts of interest.

T.R. Cox acknowledges funding support from the National Health and Medical Research Council (NHMRC) Ideas (2000937), Project (1129766, 1140125), Development (2013881) and Fellowship (1158590) schemes, a Cancer Institute NSW Career Development Fellowship (CDF171105), Cancer Council NSW project support (RG19-09, RG23-11) and Susan G. Komen for the Cure (CCR17483294).

T. R. Cox reports no competing interests.

T. A. Guise is funded by the Cancer Prevention and Research Institute of Texas Grant 00011633.

T. A. Guise reports no conflicts of interest.

M. Jamal-Hanjani is a CRUK Career Establishment Awardee and has received funding from CRUK, IASLC International Lung Cancer Foundation, Lung Cancer Research Foundation, Rosetrees Trust, UKI NETs, NIHR, NIHR UCLH Biomedical Research Centre.

M. Jamal-Hanjani reports other support from Achilles Therapeutics Scientific Advisory Board and Steering Committee, Pfizer, Astex Pharmaceuticals, Oslo Cancer Cluster, and Bristol Myers Squibb outside the submitted work.

T. Janowitz acknowledges funding from Cancer Grand Challenges (NIH: 1OT2CA278690-01; CRUK: CGCATF-2021/100019), Cancer Research UK (C42738/A24868), the Mark Foundation for Cancer Research (33300111), Cold Spring Harbor Laboratory (CSHL), and developmental funds from CSHL Cancer Center Support Grant 5P30CA045508. The CRUK CI (Li Ka Shing Centre), where some of this work was performed, was generously funded by CK Hutchison Holdings Limited, the University of Cambridge, CRUK, the Atlantic Philanthropies, and others.

T. Janowitz reports no conflicts of interest.

R. Kaplan is funded by the Intramural Research Program, the National Cancer Institute, NIH Clinical Center, and the National Institutes of Health (NIH NCI ZIABC011332-06 and NIH NCI ZIABC011334-10).

R. Kaplan reports no conflicts of interest.

R. Lee is supported by a Wellcome Early Career Investigator Award (225724/Z/22/Z).

R. Lee is on advisory board of Pierre Fabre.

E. Sahai is supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (CC2040), the UK Medical Research Council (CC2040), and the Wellcome Trust (CC2040) and the European Research Council (ERC Advanced Grant CAN_ORGANISE, Grant agreement number 101019366).

E. Sahai reports grants from Mark Foundation and the European Research Council during the conduct of the study; grants from Novartis, Merck Sharp Dohme, AstraZeneca and personal fees from Phenomic outside the submitted work.

C. Swanton is a Royal Society Napier Research Professor (RSRP\R\210001). His work is supported by the Francis Crick Institute that receives its core funding from Cancer Research UK (CC2041), the UK Medical Research Council (CC2041), and the Wellcome Trust (CC2041) and the European Research Council under the European Union’s Horizon 2020 research and innovation program (ERC Advanced Grant PROTEUS Grant agreement no. 835297).

C. Swanton reports grants and personal fees from Bristol Myers Squibb, AstraZeneca, Boehringer-Ingelheim, Roche-Ventana, personal fees from Pfizer, grants from Ono Pharmaceutical, Personalis, grants, personal fees, and other support from GRAIL, other support from AstraZeneca and GRAIL, personal fees and other support from Achilles Therapeutics, Bicycle Therapeutics, personal fees from Genentech, Medixci, China Innovation Centre of Roche (CiCoR) formerly Roche Innovation Centre, Metabomed, Relay Therapeutics, Saga Diagnostics, Sarah Canon Research Institute, Amgen, GlaxoSmithKline, Illumina, MSD, Novartis, other support from Apogen Biotechnologies and Epic Bioscience during the conduct of the study; grants and personal fees from BMS, AstraZeneca, Boehringer-Ingelheim, Roche-Ventana, grants from Ono Pharmaceuticals, Personalis, personal fees and other support from GRAIL, Achilles Therapeutics, Bicycle Therapeutics, Relay Therapeutics, personal fees from Genentech, Medixci, China Innovation Centre of Roche, Metabomed, Saga Diagnostics, Sarah Canon Research Institute, Amgen, GSK, Illumina, MSD, other support from Apogen Biosciences, and Epic Biosciences outside the submitted work; in addition, C. Swanton has a patent for PCT/ US2017/028013 licensed to Natera Inc, UCL Business, a patent for PCT/EP2016/059401 licensed to Cancer Research Technology, a patent for PCT/EP2016/071471 issued to Cancer Research Technology, a patent for PCT/GB2018/051912 pending, a patent for PCT/GB2018/052004 issued to Francis Crick Institute, University College London, Cancer Research Technology Ltd, a patent for PCT/ GB2020/050221 issued to Francis Crick Institute, University College London, a patent for PCT/EP2022/077987 pending to Cancer Research Technology, a patent for PCT/GB2017/053289 licensed, a patent for PCT/EP2022/077987 pending to Francis Crick Institute, a patent for PCT/EP2023/059039 pending to Francis Crick Institute, and a patent for PCT/GB2018/051892 pending to Francis Crick Institute. For the purpose of Open Access, the author has applied a CC BY public copyright license to any Author Accepted Manuscript version arising from this submission. C.S is Co-chief Investigator of NHS Galleri trial funded by GRAIL. He is Chief Investigator for the AstraZeneca MeRmaiD I and II clinical trials and Chair of the Steering Committee. C.S is cofounder of Achilles Therapeutics and holds stock options.

M.G. Vander Heiden reports support from the Lustgarten Foundation, the MIT Center for Precision Cancer Medicine, the Ludwig Center at MIT, and NIH grants R35 CA242379 and P30 CA1405141.

M.G. Vander Heiden is a scientific advisor for Agios Pharmaceuticals, iTeos Therapeutics, Sage Therapeutics, Faeth Therapeutics, Droia Ventures, and Auron Therapeutics on topics unrelated to the presented work.

Footnotes

Author contributions

E.S. and T.R.C collated both written and verbal input from all authors and wrote the manuscript with assistance from R.L. All authors contributed to discussion of the content and assisted with editing for accuracy and clarity. All authors reviewed and/or edited the manuscript before submission.

References

  • 1.Dillekås H, Rogers MS, Straume O. Are 90% of deaths from cancer caused by metastases? [cited 2024 Jan 21];Cancer Med. 2019 Sep 1;8(12):5574–6. doi: 10.1002/cam4.2474. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Seyfried TN, Huysentruyt LC. On the origin of cancer metastasis. [cited 2024 Jan 21];Crit Rev Oncog. 2013 18(1–2):43–73. doi: 10.1615/critrevoncog.v18.i1-2.40. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Riihimäki M, Hemminki A, Fallah M, Thomsen H, Sundquist K, Sundquist J, et al. Metastatic sites and survival in lung cancer. [cited 2024 Jan 21];Lung Cancer. 2014 Oct 1;86(1):78–84. doi: 10.1016/j.lungcan.2014.07.020. [Internet] [DOI] [PubMed] [Google Scholar]
  • 4.Wheatley-Price P, Blackhall F, Thatcher N. The influence of sex in non-small cell lung cancer. [cited 2024 Jan 21];Onkologie. 2009 Oct;32(10):547–8. doi: 10.1159/000235609. [Internet] [DOI] [PubMed] [Google Scholar]
  • 5.Abu-Sbeih H, Faleck DM, Ricciuti B, Mendelsohn RB, Naqash AR, Cohen JV, et al. Immune Checkpoint Inhibitor Therapy in Patients With Preexisting Inflammatory Bowel Disease. [cited 2024 Jan 21];Journal of Clinical Oncology. 2020 Feb 2;38(6):576. doi: 10.1200/JCO.19.01674. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Neugut AI, Matasar M, Wang X, McBride R, Jacobson JS, Tsai WY, et al. Duration of adjuvant chemotherapy for colon cancer and survival among the elderly. [cited 2024 Jan 21];J Clin Oncol. 2006 May 20;24(15):2368–75. doi: 10.1200/JCO.2005.04.5005. [Internet] [DOI] [PubMed] [Google Scholar]
  • 7.Sullivan DR, Chan B, Lapidus JA, Ganzini L, Hansen L, Carney PA, et al. Association of Early Palliative Care Use With Survival and Place of Death Among Patients With Advanced Lung Cancer Receiving Care in the Veterans Health Administration. [cited 2024 Apr 5];JAMA Oncol. 2019 Dec 1;5(12):1702–9. doi: 10.1001/jamaoncol.2019.3105. [Internet] Available from: https://jamanetwork.com/journals/jamaoncology/fullarticle/2751526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sallnow L, Smith R, Ahmedzai SH, Bhadelia A, Chamberlain C, Cong Y, et al. Report of the Lancet Commission on the Value of Death: bringing death back into life. [cited 2024 Jan 21];The Lancet. 2022 Feb 26;399(10327):837–84. doi: 10.1016/S0140-6736(21)02314-X. [Internet] Available from: http://www.thelancet.com/article/S014067362102314X/fulltext. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Abdel-Karim IA, Sammel RB, Prange MA. Causes of death at autopsy in an inpatient hospice program. [cited 2024 Apr 4];J Palliat Med. 2007 Aug;10(4):894–8. doi: 10.1089/jpm.2006.0240. [Internet] [DOI] [PubMed] [Google Scholar]
  • 10.Pautex S, Vayne-Bossert P, Jamme S, Herrmann F, Vilarino R, Weber C, et al. Anatomopathological causes of death in patients with advanced cancer: association with the use of anticoagulation and antibiotics at the end of life. [cited 2024 Apr 4];J Palliat Med. 2013 16(6):669–74. doi: 10.1089/jpm.2012.0369. [Internet] [DOI] [PubMed] [Google Scholar]
  • 11.Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH. Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. [cited 2024 Jan 21];J Thromb Haemost. 2007 Mar;5(3):632–4. doi: 10.1111/j.1538-7836.2007.02374.x. [Internet] [DOI] [PubMed] [Google Scholar]
  • 12.Levi M, Scully M. How I treat disseminated intravascular coagulation. [cited 2024 Jan 21];Blood. 2018 Feb 22;131(8):845–54. doi: 10.1182/blood-2017-10-804096. [Internet] [DOI] [PubMed] [Google Scholar]
  • 13.Anker MS, Sanz AP, Zamorano JL, Mehra MR, Butler J, Riess H, et al. Advanced cancer is also a heart failure syndrome: a hypothesis. [cited 2024 Jan 21];J Cachexia Sarcopenia Muscle. 2021 Jun 1;12(3):533. doi: 10.1002/jcsm.12694. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Asdahl PH, Sundbøll J, Adelborg K, Rasmussen TB, Seesaghur AM, Hernandez RK, et al. Cardiovascular events in cancer patients with bone metastases-A Danish population-based cohort study of 23,113 patients. [cited 2024 Jan 22];Cancer Med. 2021 Jul 1;10(14):4885–95. doi: 10.1002/cam4.4027. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sinn DH, Cho JY, Gwak GY, Paik YH, Choi MS, Lee JH, et al. Different survival of Barcelona clinic liver cancer stage C hepatocellular carcinoma patients by the extent of portal vein invasion and the type of extrahepatic spread. [cited 2024 Jan 21];PLoS One. 2015 Apr 29;10(4) doi: 10.1371/journal.pone.0124434. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zisman A, Wieder JA, Pantuck AJ, Chao DH, Dorey F, Said JW, et al. Renal cell carcinoma with tumor thrombus extension: biology, role of nephrectomy and response to immunotherapy. [cited 2024 Jan 21];J Urol. 2003 Mar 1;169(3):909–16. doi: 10.1097/01.ju.0000045706.35470.1e. [Internet] [DOI] [PubMed] [Google Scholar]
  • 17.Suárez C, Fernández-Alvarez V, Hamoir M, Mendenhall WM, Strojan P, Quer M, et al. Carotid blowout syndrome: modern trends in management. [cited 2024 Jan 21];Cancer Manag Res. 2018 10:5617. doi: 10.2147/CMAR.S180164. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lin AL, Avila EK. Neurologic Emergencies in the Cancer Patient: Diagnosis and Management. [cited 2024 Jan 21];J Intensive Care Med. 2017 Feb 1;32(2):99. doi: 10.1177/0885066615619582. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gamburg ES, Regine WF, Patchell RA, Strottmann JM, Mohiuddin M, Young AB. The prognostic significance of midline shift at presentation on survival in patients with glioblastoma multiforme. [cited 2024 Jan 21];Int J Radiat Oncol Biol Phys. 2000 Dec 1;48(5):1359–62. doi: 10.1016/s0360-3016(00)01410-3. [Internet] [DOI] [PubMed] [Google Scholar]
  • 20.Mokri B. The Monro-Kellie hypothesis: applications in CSF volume depletion. [cited 2024 Jan 21];Neurology. 2001 Jun 26;56(12):1746–8. doi: 10.1212/wnl.56.12.1746. [Internet] [DOI] [PubMed] [Google Scholar]
  • 21.Mastall M, Wolpert F, Gramatzki D, Imbach L, Becker D, Schmick A, et al. Survival of brain tumour patients with epilepsy. [cited 2024 Jan 21];Brain. 2021 Dec 16;144(11):3322–7. doi: 10.1093/brain/awab188. [Internet] [DOI] [PubMed] [Google Scholar]
  • 22.Steindl A, Yadavalli S, Gruber KA, Seiwald M, Gatterbauer B, Dieckmann K, et al. Neurological symptom burden impacts survival prognosis in patients with newly diagnosed non–small cell lung cancer brain metastases. [cited 2024 Jan 21];Cancer. 2020 Oct 1;126(19):4341–52. doi: 10.1002/cncr.33085. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Girard N, Deshpande C, Lau C, Finley D, Rusch V, Pao W, et al. Comprehensive histologic assessment helps to differentiate multiple lung primary nonsmall cell carcinomas from metastases. [cited 2024 Feb 1];Am J Surg Pathol. 2009 Dec;33(12):1752–64. doi: 10.1097/PAS.0b013e3181b8cf03. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lee P, Weerasuriya DK, Lavori PW, Quon A, Hara W, Maxim PG, et al. Metabolic Tumor Burden Predicts for Disease Progression and Death in Lung Cancer. International Journal of Radiation Oncology*Biology*Physics. 2007 Oct 1;69(2):328–33. doi: 10.1016/j.ijrobp.2007.04.036. [DOI] [PubMed] [Google Scholar]
  • 25.Lee P, Weerasuriya DK, Lavori PW, Quon A, Hara W, Maxim PG, et al. Metabolic Tumor Burden Predicts for Disease Progression and Death in Lung Cancer. International Journal of Radiation Oncology*Biology*Physics. 2007 Oct 1;69(2):328–33. doi: 10.1016/j.ijrobp.2007.04.036. [DOI] [PubMed] [Google Scholar]
  • 26.Kookoolis AS, Puchalski JT, Murphy TE, Araujo KL, Pisani MA. Mortality of Hospitalized Patients with Pleural Effusions. [cited 2024 Jan 21];J Pulm Respir Med. 2014 4(3):184. doi: 10.4172/2161-105X.1000184. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cousins SE, Tempest E, Feuer DJ. Surgery for the resolution of symptoms in malignant bowel obstruction in advanced gynaecological and gastrointestinal cancer. [cited 2024 Jan 21];Cochrane Database of Systematic Reviews. 2016 Jan 4;2016(3) doi: 10.1002/14651858.CD002764.pub2/full. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Baker ML, Yamamoto Y, Perazella MA, Dizman N, Shirali AC, Hafez N, et al. Mortality after acute kidney injury and acute interstitial nephritis in patients prescribed immune checkpoint inhibitor therapy. [cited 2024 Jan 22];J Immunother Cancer. 2022 Mar 1;10(3):e004421. doi: 10.1136/jitc-2021-004421. [Internet] Available from: https://jitc.bmj.com/content/10/3/e004421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bhave P, Buckle A, Sandhu S, Sood S. Mortality due to immunotherapy related hepatitis. [cited 2024 Jan 22];J Hepatol. 2018 Oct 1;69(4):976–8. doi: 10.1016/j.jhep.2018.06.012. [Internet] Available from: http://www.journal-of-hepatology.eu/article/S0168827818321652/fulltext. [DOI] [PubMed] [Google Scholar]
  • 30.Lameire NH, Flombaum CD, Moreau D, Ronco C. Acute renal failure in cancer patients. [cited 2024 Jan 22];Ann Med. 2005 37(1):13–25. doi: 10.1080/07853890510007205. [Internet] [DOI] [PubMed] [Google Scholar]
  • 31.Ries F, Klastersky J. Nephrotoxicity induced by cancer chemotherapy with special emphasis on cisplatin toxicity. [cited 2024 Jan 22];Am J Kidney Dis. 1986 8(5):368–79. doi: 10.1016/s0272-6386(86)80112-3. [Internet] [DOI] [PubMed] [Google Scholar]
  • 32.Wong JL, Evans SE. Bacterial Pneumonia in Patients with Cancer: Novel Risk Factors and Management. Clin Chest Med. 2017 Jun 1;38(2):263–77. doi: 10.1016/j.ccm.2016.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee LYW, Cazier JB, Angelis V, Arnold R, Bisht V, Campton NA, et al. COVID-19 mortality in patients with cancer on chemotherapy or other anticancer treatments: a prospective cohort study. [cited 2024 Jan 22];Lancet. 2020 Jun 20;395(10241):1919–26. doi: 10.1016/S0140-6736(20)31173-9. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Williamson EJ, Walker AJ, Bhaskaran K, Bacon S, Bates C, Morton CE, et al. Factors associated with COVID-19-related death using OpenSAFELY. [cited 2020 Jul 8];Nature. 2020 Jul 8;584(7821):430–6. doi: 10.1038/s41586-020-2521-4. [Internet] Available from: http://www.nature.com/articles/s41586-020-2521-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Donovan PJ, Achong N, Griffin K, Galligan J, Pretorius CJ, McLeod DSA. PTHrP-mediated hypercalcemia: causes and survival in 138 patients. [cited 2024 Jan 22];J Clin Endocrinol Metab. 2015 May 1;100(5):2024–9. doi: 10.1210/jc.2014-4250. [Internet] [DOI] [PubMed] [Google Scholar]
  • 36.Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Ito S, Ogawa Y, et al. Diagnosis and management of insulinoma. [cited 2024 Jan 22];World J Gastroenterol. 2013 19(6):829–37. doi: 10.3748/wjg.v19.i6.829. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Burtis WJ, Brady TG, Orloff JJ, Ersbak JB, Warrell RP, Olson BR, et al. Immunochemical characterization of circulating parathyroid hormone-related protein in patients with humoral hypercalcemia of cancer. [cited 2024 Jan 22];N Engl J Med. 1990 Apr 19;322(16):1106–12. doi: 10.1056/NEJM199004193221603. [Internet] [DOI] [PubMed] [Google Scholar]
  • 38.Ellison DH, Berl T. The Syndrome of Inappropriate Antidiuresis. [cited 2024 Jan 22];2007 May 17;356(20):2064–72. doi: 10.1056/NEJMcp066837. [Internet] Available from: https://www.nejm.org/doi/full/10.1056/NEJMcp066837. [DOI] [PubMed] [Google Scholar]
  • 39.Giometto B, Grisold W, Vitaliani R, Graus F, Honnorat J, Bertolini G, et al. Paraneoplastic Neurologic Syndrome in the PNS Euronetwork Database: A European Study From 20 Centers. [cited 2024 Jan 22];Arch Neurol. 2010 Mar 1;67(3):330–5. doi: 10.1001/archneurol.2009.341. [Internet] Available from: https://jamanetwork.com/journals/jamaneurology/fullarticle/799603. [DOI] [PubMed] [Google Scholar]
  • 40.Pelosof LC, Gerber DE. Paraneoplastic syndromes: an approach to diagnosis and treatment. [cited 2024 Jan 22];Mayo Clin Proc. 2010 85(9):838–54. doi: 10.4065/mcp.2010.0099. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. [cited 2024 Jan 22];JAMA Oncol. 2018 Dec 1;4(12):1721. doi: 10.1001/jamaoncol.2018.3923. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Feng S, Coward J, McCaffrey E, Coucher J, Kalokerinos P, O’Byrne K. Pembrolizumab-Induced Encephalopathy: A Review of Neurological Toxicities with Immune Checkpoint Inhibitors. [cited 2024 Apr 5];Journal of Thoracic Oncology. 2017 Nov 1;12(11):1626–35. doi: 10.1016/j.jtho.2017.08.007. [Internet] Available from: http://www.jto.org/article/S1556086417306822/fulltext. [DOI] [PubMed] [Google Scholar]
  • 43.Coustal C, Vanoverschelde J, Quantin X, Lesage C, Michot JM, Lappara A, et al. Prognosis of immune checkpoint inhibitors-induced myocarditis: a case series. [cited 2024 Apr 5];J Immunother Cancer. 2023 May 1;11(5):e004792. doi: 10.1136/jitc-2022-004792. [Internet] Available from: https://jitc.bmj.com/content/11/5/e004792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kuderer NM, Dale DC, Crawford J, Cosler LE, Lyman GH. Mortality, morbidity, and cost associated with febrile neutropenia in adult cancer patients. [cited 2024 Jan 22];Cancer. 2006 May 15;106(10):2258–66. doi: 10.1002/cncr.21847. [Internet] [DOI] [PubMed] [Google Scholar]
  • 45.Ghanavat M, Ebrahimi M, Rafieemehr H, Maniati M, Behzad MM, Shahrabi S. Thrombocytopenia in solid tumors: Prognostic significance. [cited 2024 Jan 31];Oncol Rev. 2019 13(1):43–8. doi: 10.4081/oncol.2019.413. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Agarwal MA, Sridharan A, Pimentel RC, Markowitz SM, Rosenfeld LE, Fradley MG, et al. Ventricular Arrhythmia in Cancer Patients: Mechanisms, Treatment Strategies and Future Avenues. [cited 2024 Jan 22];Arrhythm Electrophysiol Rev. 2023 12 doi: 10.15420/aer.2023.04. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zafar A, Drobni ZD, Mosarla R, Alvi RM, Lei M, Lou UY, et al. The Incidence, Risk Factors, and Outcomes With 5-Fluorouracil–Associated Coronary Vasospasm. JACC CardioOncol. 2021 Mar 1;3(1):101–9. doi: 10.1016/j.jaccao.2020.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Polk A, Shahmarvand N, Vistisen K, Vaage-Nilsen M, Larsen FO, Schou M, et al. Incidence and risk factors for capecitabine-induced symptomatic cardiotoxicity: a retrospective study of 452 consecutive patients with metastatic breast cancer. [cited 2024 Jan 22];BMJ Open. 2016 Oct 1;6(10):e012798. doi: 10.1136/bmjopen-2016-012798. [Internet] Available from: https://bmjopen.bmj.com/content/6/10/e012798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Safdar A, Bodey G, Armstrong D. Infections in Patients with Cancer: Overview. [cited 2024 Jan 22];Principles and Practice of Cancer Infectious Diseases. 2011 3 [Internet] [Google Scholar]
  • 50.Foster DS, Jones RE, Ransom RC, Longaker MT, Norton JA. The evolving relationship of wound healing and tumor stroma. [cited 2024 Jan 23];JCI Insight. 2018 Sep 9;3(18) doi: 10.1172/jci.insight.99911. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Park SJ, Bejar R. Clonal Hematopoiesis in Cancer. [cited 2024 Jan 23];Exp Hematol. 2020 Mar 1;83:105. doi: 10.1016/j.exphem.2020.02.001. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Liebman HA. Thrombocytopenia in cancer patients. [cited 2024 Jan 23];Thromb Res. 2014 133(Suppl 2) doi: 10.1016/S0049-3848(14)50011-4. [Internet] (SUPPL. 2) [DOI] [PubMed] [Google Scholar]
  • 53.Chakraborty R, Rybicki L, Nakashima MO, Dean RM, Faiman BM, Samaras CJ, et al. Characterisation and prognostic impact of immunoparesis in relapsed multiple myeloma. [cited 2024 Jan 22];Br J Haematol. 2020 Jun 1;189(6):1074–82. doi: 10.1111/bjh.16488. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Allen BM, Hiam KJ, Burnett CE, Venida A, DeBarge R, Tenvooren I, et al. Systemic dysfunction and plasticity of the immune macroenvironment in cancer models. [cited 2024 Jan 23];Nature Medicine. 2020 May 25;26(7):1125–34. doi: 10.1038/s41591-020-0892-6. 2020 26:7 [Internet] Available from: https://www.nature.com/articles/s41591-020-0892-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Munn DH, Bronte V. Immune suppressive mechanisms in the tumor microenvironment. Curr Opin Immunol. 2016 Apr 1;39:1–6. doi: 10.1016/j.coi.2015.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kochar R, Banerjee S. Infections of the biliary tract. [cited 2024 Jan 22];Gastrointest Endosc Clin N Am. 2013 Apr;23(2):199–218. doi: 10.1016/j.giec.2012.12.008. [Internet] [DOI] [PubMed] [Google Scholar]
  • 57.Valvani A, Martin A, Devarajan A, Chandy D. Postobstructive pneumonia in lung cancer. [cited 2024 Jan 22];Ann Transl Med. 2019 Aug;7(15):357. doi: 10.21037/atm.2019.05.26. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rolston KVI. Infections in Cancer Patients with Solid Tumors: A Review. [cited 2024 Jan 22];Infect Dis Ther. 2017 Mar 1;6(1):69–83. doi: 10.1007/s40121-017-0146-1. [Internet] Available from: https://link.springer.com/article/10.1007/s40121-017-0146-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wu X, Li Z, Cao J, Jiao J, Wang Y, Liu G, et al. The association between major complications of immobility during hospitalization and quality of life among bedridden patients: A 3 month prospective multi-center study. [cited 2024 Jan 22];PLoS One. 2018 Oct 1;13(10) doi: 10.1371/journal.pone.0205729. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kasthuri RS, Taubman MB, Mackman N. Role of Tissue Factor in Cancer. [cited 2024 Jan 22];Journal of Clinical Oncology. 2009 Oct 10;27(29):4834. doi: 10.1200/JCO.2009.22.6324. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wade JC. Viral Infections in Patients with Hematological Malignancies. [cited 2024 Jan 22];Hematology. 2006 Jan 1;2006(1):368–74. doi: 10.1182/asheducation-2006.1.368. [Internet] [DOI] [PubMed] [Google Scholar]
  • 62.Ersvaer E, Liseth K, Skavland J, Gjertsen BT, Bruserud Ø. Intensive chemotherapy for acute myeloid leukemia differentially affects circulating TC1, TH1, TH17 and TREGcells. [cited 2024 Jan 22];BMC Immunol. 2010 Jul 9;11(1):1–12. doi: 10.1186/1471-2172-11-38. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kuter DJ. Treatment of chemotherapy-induced thrombocytopenia in patients with non-hematologic malignancies. [cited 2024 Jan 23];Haematologica. 2022 Jun 6;107(6):1243. doi: 10.3324/haematol.2021.279512. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Rodgers GM, Becker PS, Blinder M, Cella D, Chanan-Khan A, Cleeland C, et al. Cancer- and chemotherapy-induced anemia. [cited 2024 Jan 23];J Natl Compr Canc Netw. 2012 May 1;10(5):628–53. doi: 10.6004/jnccn.2012.0064. [Internet] [DOI] [PubMed] [Google Scholar]
  • 65.Nesher L, Rolston KVI. The current spectrum of infection in cancer patients with chemotherapy related neutropenia. [cited 2024 Jan 23];Infection. 2014 Aug 23;42(1):5–13. doi: 10.1007/s15010-013-0525-9. [Internet] Available from: https://link.springer.com/article/10.1007/s15010-013-0525-9. [DOI] [PubMed] [Google Scholar]
  • 66.Blijlevens NMA, Logan RM, Netea MG. Mucositis: from febrile neutropenia to febrile mucositis. [cited 2024 Jan 23];Journal of Antimicrobial Chemotherapy. 2009 May 1;63(Suppl_1):i36–40. doi: 10.1093/jac/dkp081. [Internet] [DOI] [PubMed] [Google Scholar]
  • 67.Petrelli F, Bukovec R, Perego G, Luisa R, Luciani A, Zaniboni A, et al. Association of steroid use with survival in solid tumours. Eur J Cancer. 2020 Dec 1;141:105–14. doi: 10.1016/j.ejca.2020.09.020. [DOI] [PubMed] [Google Scholar]
  • 68.Rolston KVI. Infections in Cancer Patients with Solid Tumors: A Review. [cited 2024 Jan 22];Infect Dis Ther. 2017 Mar 1;6(1):69–83. doi: 10.1007/s40121-017-0146-1. [Internet] Available from: https://link.springer.com/article/10.1007/s40121-017-0146-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bhatia R, Holtan S, El Jurdi N, Prizment A, Blaes A. Do Cancer and Cancer Treatments Accelerate Aging? [cited 2024 Apr 14];Curr Oncol Rep. 2022 Nov 1;24(11):1401. doi: 10.1007/s11912-022-01311-2. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Eisenstein TK. The Role of Opioid Receptors in Immune System Function. [cited 2024 Apr 4];Front Immunol. 2019 Dec 20;10:485158. doi: 10.3389/fimmu.2019.02904. [Internet] Available from: www.frontiersin.org. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Böll B, Schalk E, Buchheidt D, Hasenkamp J, Kiehl M, Kiderlen TR, et al. Central venous catheter–related infections in hematology and oncology: 2020 updated guidelines on diagnosis, management, and prevention by the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO. [cited 2024 Jan 22];Ann Hematol. 2021 Jan 1;100(1):239. doi: 10.1007/s00277-020-04286-x. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ruiz-Giardin JM, Ochoa Chamorro I, Velázquez Riós L, Jaqueti Aroca J, Garciá Arata MI, Sanmartín López JV, et al. Blood stream infections associated with central and peripheral venous catheters. [cited 2024 Jan 22];BMC Infectious Diseases. 2019 Oct 15;19(1):1–9. doi: 10.1186/s12879-019-4505-2. 2019 19:1 [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. Current concepts in the diagnosis and management of cytokine release syndrome. [cited 2024 Jan 22];Blood. 2014 Jul 10;124(2):188–95. doi: 10.1182/blood-2014-05-552729. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Brahmer JR, Long GV, Hamid O, Garon EB, Herbst RS, Andre T, et al. Safety profile of pembrolizumab monotherapy based on an aggregate safety evaluation of 8937 patients. [cited 2024 Jan 22];Eur J Cancer. 2024 Jan 11;:113530. doi: 10.1016/j.ejca.2024.113530. [Internet] Available from: https://linkinghub.elsevier.com/retrieve/pii/S0959804924000066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. [cited 2024 Jan 22];New England Journal of Medicine. 2019 Oct 17;381(16):1535–46. doi: 10.1056/nejmoa1910836. [Internet] [DOI] [PubMed] [Google Scholar]
  • 76.Vozy A, De Martin E, Johnson DB, Lebrun-Vignes B, Moslehi JJ, Salem JE. Increased reporting of fatal hepatitis associated with immune checkpoint inhibitors. Eur J Cancer. 2019 Dec 1;123:112–5. doi: 10.1016/j.ejca.2019.09.022. [DOI] [PubMed] [Google Scholar]
  • 77.Palaskas N, Lopez-Mattei J, Durand JB, Iliescu C, Deswal A. Immune Checkpoint Inhibitor Myocarditis: Pathophysiological Characteristics, Diagnosis, and Treatment. [cited 2024 Jan 22];J Am Heart Assoc. 2020 Jan 21;9(2) doi: 10.1161/JAHA.119.013757. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Janssen JBE, Leow TYS, Herbschleb KH, Gijtenbeek JMM, Boers-Sonderen MJ, Gerritsen WR, et al. Immune Checkpoint Inhibitor-related Guillain-Barré Syndrome: A Case Series and Review of the Literature. [cited 2024 Jan 22];J Immunother. 2021 Sep 1;44(7):276–82. doi: 10.1097/CJI.0000000000000364. [Internet] [DOI] [PubMed] [Google Scholar]
  • 79.Camelliti S, Le Noci V, Bianchi F, Moscheni C, Arnaboldi F, Gagliano N, et al. Mechanisms of hyperprogressive disease after immune checkpoint inhibitor therapy: what we (don’t) know. [cited 2024 Apr 5];J Exp Clin Cancer Res. 2020 Dec 1;39(1) doi: 10.1186/s13046-020-01721-9. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kitamura W, Asada N, Naoi Y, Abe M, Fujiwara H, Ennishi D, et al. Bone marrow microenvironment disruption and sustained inflammation with prolonged haematologic toxicity after CAR T-cell therapy. [cited 2024 Jan 22];Br J Haematol. 2023 Jul 1;202(2):294–307. doi: 10.1111/bjh.18747. [Internet] [DOI] [PubMed] [Google Scholar]
  • 81.Seano G, Nia HT, Emblem KE, Datta M, Ren J, Krishnan S, et al. Solid stress in brain tumours causes neuronal loss and neurological dysfunction and can be reversed by lithium. [cited 2024 Jan 22];Nat Biomed Eng. 2019 Mar 1;3(3):230. doi: 10.1038/s41551-018-0334-7. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Madhusoodanan S, Ting MB, Farah T, Ugur U. Psychiatric aspects of brain tumors: A review. [cited 2024 Jan 22];World J Psychiatry. 2015 Sep 9;5(3):273. doi: 10.5498/wjp.v5.i3.273. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Gerstenecker A, Nabors LB, Meneses K, Fiveash JB, Marson DC, Cutter G, et al. Cognition in patients with newly diagnosed brain metastasis: profiles and implications. [cited 2024 Jan 22];J Neurooncol. 2014 Sep 27;120(1):179. doi: 10.1007/s11060-014-1543-x. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Krishna S, Choudhury A, Keough MB, Seo K, Ni L, Kakaizada S, et al. Glioblastoma remodelling of human neural circuits decreases survival. [cited 2024 Jan 22];Nature. 2023 May 3;617(7961):599–607. doi: 10.1038/s41586-023-06036-1. 2023 617:7961 [Internet] Available from: https://www.nature.com/articles/s41586-023-06036-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Taylor KR, Barron T, Hui A, Spitzer A, Yalçin B, Ivec AE, et al. Glioma synapses recruit mechanisms of adaptive plasticity. [cited 2024 Jan 22];Nature. 2023 Nov 1;623(7986):366–74. doi: 10.1038/s41586-023-06678-1. 2023 623:7986 [Internet] Available from: https://www.nature.com/articles/s41586-023-06678-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hanahan D, Monje M. Cancer hallmarks intersect with neuroscience in the tumor microenvironment. [cited 2024 Jan 22];Cancer Cell. 2023 41:573–80. doi: 10.1016/j.ccell.2023.02.012. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ahles TA, Root JC. Cognitive Effects of Cancer and Cancer Treatments. [cited 2024 Jan 22];2018 May 7;14:425–51. doi: 10.1146/annurev-clinpsy-050817-084903. [Internet] Available from: https://www.annualreviews.org/doi/abs/10.1146/annurev-clinpsy-050817-084903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Allexandre D, Seyidova-Khoshknabi D, Davis MP, Ranganathan VK, Siemionow V, Walsh D, et al. EEG Correlates of Central Origin of Cancer-Related Fatigue. [cited 2024 Jan 23];Neural Plast. 2020 2020 doi: 10.1155/2020/8812984. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Büttner-Teleagă A, Kim YT, Osel T, Richter K. Sleep Disorders in Cancer-A Systematic Review. [cited 2024 Jan 22];Int J Environ Res Public Health. 2021 Nov 1;18(21) doi: 10.3390/ijerph182111696. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Walsh D, Nelson KA. Autonomic nervous system dysfunction in advanced cancer. [cited 2024 Feb 1];Supportive Care in Cancer. 2002 Feb 23;10(7):523–8. doi: 10.1007/s00520-002-0376-x. [Internet] [DOI] [PubMed] [Google Scholar]
  • 91.Ghandour F, Squassina A, Karaky R, Diab-Assaf M, Fadda P, Pisanu C. Presenting Psychiatric and Neurological Symptoms and Signs of Brain Tumors before Diagnosis: A Systematic Review. [cited 2024 Jan 22];Brain Sci. 2021 Mar 1;11(3):1–20. doi: 10.3390/brainsci11030301. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Akechi T, Nakano T, Akizuki N, Okamura M, Sakuma K, Nakanishi T, et al. Somatic Symptoms for Diagnosing Major Depression in Cancer Patients. Psychosomatics. 2003 May 1;44(3):244–8. doi: 10.1176/appi.psy.44.3.244. [DOI] [PubMed] [Google Scholar]
  • 93.Nho JH, Kim SR, Kwon YS. Depression and appetite: predictors of malnutrition in gynecologic cancer. [cited 2024 Jan 22];Supportive Care in Cancer. 2014 Oct 3;22(11):3081–8. doi: 10.1007/s00520-014-2340-y. [Internet] [DOI] [PubMed] [Google Scholar]
  • 94.Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, et al. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. [cited 2024 Apr 5];Nat Med. 2006 12(8):939–44. doi: 10.1038/nm1447. [Internet] [DOI] [PubMed] [Google Scholar]
  • 95.Chang A, Botteri E, Gillis RD, Löfling L, Le CP, Ziegler AI, et al. Beta-blockade enhances anthracycline control of metastasis in triple-negative breast cancer. [cited 2024 Apr 5];Sci Transl Med. 2023 15(693) doi: 10.1126/scitranslmed.adf1147. [Internet] [DOI] [PubMed] [Google Scholar]
  • 96.Magnon C, Hall SJ, Lin J, Xue X, Gerber L, Freedland SJ, et al. Autonomic nerve development contributes to prostate cancer progression. [cited 2024 Apr 5];Science. 2013 341(6142) doi: 10.1126/science.1236361. [Internet] [DOI] [PubMed] [Google Scholar]
  • 97.Bowden MB, Walsh NJ, Jones AJ, Talukder AM, Lawson AG, Kruse EJ. Demographic and clinical factors associated with suicide in gastric cancer in the United States. [cited 2024 Apr 5];J Gastrointest Oncol. 2017 Oct 1;8(5):897–901. doi: 10.21037/jgo.2017.08.02. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pinquart M, Duberstein PR. Depression and cancer mortality: a meta-analysis. [cited 2024 Jan 22];Psychol Med. 2010 Nov;40(11):1797–810. doi: 10.1017/S0033291709992285. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Fitzgerald P, Lo C, Li M, Gagliese L, Zimmermann C, Rodin G. The relationship between depression and physical symptom burden in advanced cancer. [cited 2024 Jan 22];BMJ Support Palliat Care. 2015 Dec 1;5(4):381–8. doi: 10.1136/bmjspcare-2012-000380. [Internet] Available from: https://spcare.bmj.com/content/5/4/381. [DOI] [PubMed] [Google Scholar]
  • 100.Chida Y, Hamer M, Wardle J, Steptoe A. Do stress-related psychosocial factors contribute to cancer incidence and survival? [cited 2024 Apr 5];Nature Clinical Practice Oncology. 2008 May 20;5(8):466–75. doi: 10.1038/ncponc1134. 2008 5:8 [Internet] Available from: https://www.nature.com/articles/ncponc1134. [DOI] [PubMed] [Google Scholar]
  • 101.He XY, Gao Y, Ng D, Michalopoulou E, George S, Adrover JM, et al. Chronic stress increases metastasis via neutrophil-mediated changes to the microenvironment. [cited 2024 Apr 5];Cancer Cell. 2024 Mar 11;42(3):474–486.:e12. doi: 10.1016/j.ccell.2024.01.013. [Internet] Available from: http://www.cell.com/article/S1535610824000370/fulltext. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Fann JR, Ell K, Sharpe M. Integrating psychosocial care into cancer services. [cited 2024 Apr 5];J Clin Oncol. 2012 Apr 10;30(11):1178–86. doi: 10.1200/JCO.2011.39.7398. [Internet] [DOI] [PubMed] [Google Scholar]
  • 103.Jacobsen PB, Wagner LI. A new quality standard: the integration of psychosocial care into routine cancer care. [cited 2024 Apr 5];J Clin Oncol. 2012 Apr 10;30(11):1154–9. doi: 10.1200/JCO.2011.39.5046. [Internet] [DOI] [PubMed] [Google Scholar]
  • 104.Gorin SS, Krebs P, Badr H, Janke EA, Jim HSL, Spring B, et al. Meta-analysis of psychosocial interventions to reduce pain in patients with cancer. [cited 2024 Apr 5];J Clin Oncol. 2012 Feb 10;30(5):539–47. doi: 10.1200/JCO.2011.37.0437. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Li M, Kennedy EB, Byrne N, Gérin-Lajoie C, Katz MR, Keshavarz H, et al. Systematic review and meta-analysis of collaborative care interventions for depression in patients with cancer. [cited 2024 Apr 5];Psychooncology. 2017 May 1;26(5):573–87. doi: 10.1002/pon.4286. [Internet] [DOI] [PubMed] [Google Scholar]
  • 106.Zaorsky NG, Zhang Y, Tuanquin L, Bluethmann SM, Park HS, Chinchilli VM. Suicide among cancer patients. [cited 2024 Jan 22];Nature Communications. 2019 Jan 14;10(1):1–7. doi: 10.1038/s41467-018-08170-1. 2019 10:1 [Internet] Available from: https://www.nature.com/articles/s41467-018-08170-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hu X, Ma J, Jemal A, Zhao J, Nogueira L, Ji X, et al. Suicide Risk Among Individuals Diagnosed With Cancer in the, US., 2000-2016. [cited 2024 Jan 22];JAMA Netw Open. 2023 Jan 3;6(1):e2251863. doi: 10.1001/jamanetworkopen.2022.51863. [Internet] Available from: https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2800688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. [cited 2024 Jan 22];Nat Rev Dis Primers. 2018 Jan 18;4 doi: 10.1038/nrdp.2017.105. [Internet] [DOI] [PubMed] [Google Scholar]
  • 109.Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. [cited 2024 Jan 22];Lancet Oncol. 2011 May;12(5):489–95. doi: 10.1016/S1470-2045(10)70218-7. [Internet] [DOI] [PubMed] [Google Scholar]
  • 110.Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. [cited 2024 Jan 22];Nature Reviews Disease Primers. 2018 Jan 18;4(1):1–18. doi: 10.1038/nrdp.2017.105. 2018 4:1 [Internet] Available from: https://www.nature.com/articles/nrdp2017105. [DOI] [PubMed] [Google Scholar]
  • 111.Farkas J, von Haehling S, Kalantar-Zadeh K, Morley JE, Anker SD, Lainscak M. Cachexia as a major public health problem: frequent, costly, and deadly. [cited 2024 Jan 22];J Cachexia Sarcopenia Muscle. 2013 4(3):173–8. doi: 10.1007/s13539-013-0105-y. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Dennison EM, Sayer AA, Cooper C. Epidemiology of sarcopenia and insight into possible therapeutic targets. [cited 2024 Jan 23];Nature Reviews Rheumatology. 2017 May 4;13(6):340–7. doi: 10.1038/nrrheum.2017.60. 2017 13:6 [Internet] Available from: https://www.nature.com/articles/nrrheum.2017.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Farasat M, Watters A, Bendelow T, Schuller J, Mehler PS, Krantz MJ. Long-term cardiac arrhythmia and chronotropic evaluation in patients with severe anorexia nervosa (LACE-AN): A pilot study. [cited 2024 Apr 5];J Cardiovasc Electrophysiol. 2020 Feb 1;31(2):432–9. doi: 10.1111/jce.14338. [Internet] [DOI] [PubMed] [Google Scholar]
  • 114.Mehler PS, Anderson K, Bauschka M, Cost J, Farooq A. Emergency room presentations of people with anorexia nervosa. [cited 2024 Apr 5];J Eat Disord. 2023 Dec 1;11(1) doi: 10.1186/s40337-023-00742-x. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bourke CD, Berkley JA, Prendergast AJ. Immune Dysfunction as a Cause and Consequence of Malnutrition. [cited 2024 Jan 22];Trends Immunol. 2016 Jun 1;37(6):386–98. doi: 10.1016/j.it.2016.04.003. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tisdale MJ. Biology of Cachexia. [cited 2024 Jan 23];JNCI: Journal of the National Cancer Institute. 1997 Dec 3;89(23):1763–73. doi: 10.1093/jnci/89.23.1763. [Internet] [DOI] [PubMed] [Google Scholar]
  • 117.Babic A, Rosenthal MH, Sundaresan TK, Khalaf N, Lee V, Brais LK, et al. Adipose tissue and skeletal muscle wasting precede clinical diagnosis of pancreatic cancer. [cited 2024 Jan 22];Nat Commun. 2023 Dec 1;14(1) doi: 10.1038/s41467-023-40024-3. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Waning DL, Mohammad KS, Reiken S, Xie W, Andersson DC, John S, et al. Excess TGF-β mediates muscle weakness associated with bone metastases in mice. [cited 2024 Jan 22];Nat Med. 2015 Nov 1;21(11):1262. doi: 10.1038/nm.3961. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Greco SH, Tomkötter L, Vahle AK, Rokosh R, Avanzi A, Mahmood SK, et al. TGF-β Blockade Reduces Mortality and Metabolic Changes in a Validated Murine Model of Pancreatic Cancer Cachexia. [cited 2024 Jan 22];PLoS One. 2015 Jul 14;10(7):e0132786. doi: 10.1371/journal.pone.0132786. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Johnen H, Lin S, Kuffner T, Brown DA, Tsai VWW, Bauskin AR, et al. Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1. [cited 2024 Jan 22];Nat Med. 2007 Nov;13(11):1333–40. doi: 10.1038/nm1677. [Internet] [DOI] [PubMed] [Google Scholar]
  • 121.Al-Sawaf O, Weiss J, Skrzypski M, Lam JM, Karasaki T, Zambrana F, et al. Body composition and lung cancer-associated cachexia in TRACERx. [cited 2024 Jan 22];Nat Med. 2023 Apr 12;29(4):846–58. doi: 10.1038/s41591-023-02232-8. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Rebbapragada A, Benchabane H, Wrana JL, Celeste AJ, Attisano L. Myostatin Signals through a Transforming Growth Factor β-Like Signaling Pathway To Block Adipogenesis. [cited 2024 Jan 23];Mol Cell Biol. 2003 Oct 1;23(20):7230. doi: 10.1128/MCB.23.20.7230-7242.2003. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Queiroz AL, Dantas E, Ramsamooj S, Murthy A, Ahmed M, Zunica ERM, et al. Blocking ActRIIB and restoring appetite reverses cachexia and improves survival in mice with lung cancer. [cited 2024 Jan 23];Nature Communications. 2022 Aug 8;13(1):1–17. doi: 10.1038/s41467-022-32135-0. 2022 13:1 [Internet] Available from: https://www.nature.com/articles/s41467-022-32135-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Loumaye A, De Barsy M, Nachit M, Lause P, Frateur L, Van Maanen A, et al. Role of Activin A and Myostatin in Human Cancer Cachexia. [cited 2024 Jan 23];J Clin Endocrinol Metab. 2015 May 1;100(5):2030–8. doi: 10.1210/jc.2014-4318. [Internet] [DOI] [PubMed] [Google Scholar]
  • 125.Waning DL, Mohammad KS, Reiken S, Xie W, Andersson DC, John S, et al. Excess TGF-β mediates muscle weakness associated with bone metastases in mice. [cited 2024 Jan 22];Nat Med. 2015 Nov 1;21(11):1262–71. doi: 10.1038/nm.3961. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Greco SH, Tomkötter L, Vahle AK, Rokosh R, Avanzi A, Mahmood SK, et al. TGF-β Blockade Reduces Mortality and Metabolic Changes in a Validated Murine Model of Pancreatic Cancer Cachexia. [cited 2024 Jan 22];PLoS One. 2015 Jul 14;10(7):e0132786. doi: 10.1371/journal.pone.0132786. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Barton BE, Murphy TF. Cancer cachexia is mediated in part by the induction of IL-6-like cytokines from the spleen. [cited 2024 Jan 22];Cytokine. 2001 Dec 21;16(6):251–7. doi: 10.1006/cyto.2001.0968. [Internet] [DOI] [PubMed] [Google Scholar]
  • 128.Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and Skeletal Muscle Wasting During Cachexia. Front Physiol. 2020 Nov 19;11:597675. doi: 10.3389/fphys.2020.597675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Strassmann G, Masui Y, Chizzonite R, Fong M. Mechanisms of Experimental Cancer Cachexia Local Involvement of 11-1 in Colon-26 Tumor. 1993;150(6):2341–5. [PubMed] [Google Scholar]
  • 130.Cancer Research. American Association for Cancer Research; [cited 2024 Jan 22]. Cachectin/Tumor Necrosis Factor: A Possible Mediator of Cancer Anorexia in the Rat1. [Internet] Available from: https://aacrjournals.org/cancerres/article/48/16/4567/492924/Cachectin-Tumor-Necrosis-Factor-A-Possible. [Google Scholar]
  • 131.Wyke SM, Tisdale MJ. NF-κB mediates proteolysis-inducing factor induced protein degradation and expression of the ubiquitin–proteasome system in skeletal muscle. [cited 2024 Jan 22];Br J Cancer. 2005 Feb 2;92(4):711. doi: 10.1038/sj.bjc.6602402. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Cai D, Frantz JD, Tawa NE, Melendez PA, Oh BC, Lidov HGW, et al. IKKβ/NF-κB Activation Causes Severe Muscle Wasting in Mice. Cell. 2004 Oct 15;119(2):285–98. doi: 10.1016/j.cell.2004.09.027. [DOI] [PubMed] [Google Scholar]
  • 133.Patel HJ, Patel BM. TNF-α and cancer cachexia: Molecular insights and clinical implications. Life Sci. 2017 Feb 1;170:56–63. doi: 10.1016/j.lfs.2016.11.033. [DOI] [PubMed] [Google Scholar]
  • 134.Mergenthaler P, Lindauer U, Dienel GA, Meisel A. Sugar for the brain: the role of glucose in physiological and pathological brain function. [cited 2024 Jan 22];Trends Neurosci. 2013 Oct;36(10):587. doi: 10.1016/j.tins.2013.07.001. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Sillos EM, Shenep JL, Burghen GA, Pui CH, Behm FG, Sandlund JT. Lactic Acidosis: A Metabolic Complication of Hematologic Malignancies. Case Report and Review of the Literature. [cited 2024 Apr 5];2000 doi: 10.1002/1097-0142. [DOI] [PubMed] [Google Scholar]
  • 136.Rampello E, Fricia T, Malaguarnera M. The management of tumor lysis syndrome. [cited 2024 Apr 5]; [cited 2024 Apr 5];Nature Clinical Practice Oncology. 2006 Aug;3(8):438–47. doi: 10.1038/ncponc0581. 2006 3:8 [Internet] Available from: https://www.nature.com/articles/ncponc0581. [DOI] [PubMed] [Google Scholar]
  • 137.Delano MJ, Moldawer LL. The origins of cachexia in acute and chronic inflammatory diseases. [cited 2024 Jan 22];Nutr Clin Pract. 2006 21(1):68–81. doi: 10.1177/011542650602100168. [Internet] [DOI] [PubMed] [Google Scholar]
  • 138.Lombardi A, Villa S, Castelli V, Bandera A, Gori A. T-Cell Exhaustion in Mycobacterium tuberculosis and Nontuberculous Mycobacteria Infection: Pathophysiology and Therapeutic Perspectives. [cited 2024 Jan 22];Microorganisms. 2021 Dec 1;9(12) doi: 10.3390/microorganisms9122460. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Moldawer LL, Sattler FR. Human immunodeficiency virus-associated wasting and mechanisms of cachexia associated with inflammation. [cited 2024 Jan 22];Semin Oncol. 1998 Feb 1;25(1 Suppl 1):73–81. [Internet] Available from: https://europepmc.org/article/med/9482543. [PubMed] [Google Scholar]
  • 140.von Kobbe C. Targeting senescent cells: approaches, opportunities, challenges. [cited 2024 Jan 23];Aging (Albany NY) 2019 Dec 12;11(24):12844. doi: 10.18632/aging.102557. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Shafqat S, Chicas EA, Shafqat A, Hashmi SK. The Achilles’ heel of cancer survivors: fundamentals of accelerated cellular senescence. [cited 2024 Jan 23];J Clin Invest. 2022 Jul 7;132(13) doi: 10.1172/JCI158452. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Wang L, Lankhorst L, Bernards R. Exploiting senescence for the treatment of cancer. [cited 2024 Jan 23];Nature Reviews Cancer. 2022 Mar 3;22(6):340–55. doi: 10.1038/s41568-022-00450-9. 2022 22:6 [Internet] Available from: https://www.nature.com/articles/s41568-022-00450-9. [DOI] [PubMed] [Google Scholar]
  • 143.Bova GS, Eltoum IA, Kiernan JA, Siegal GP, Frost AR, Best CJM, et al. Optimal molecular profiling of tissue and tissue components: defining the best processing and microdissection methods for biomedical applications. [cited 2024 Feb 1];Mol Biotechnol. 2005 Feb;29(2):119–52. doi: 10.1385/MB:29:2:119. [Internet] [DOI] [PubMed] [Google Scholar]
  • 144.Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JMC, Papaemmanuil E, et al. The evolutionary history of lethal metastatic prostate cancer. [cited 2024 Feb 1];Nature. 2015 Apr 15;520(7547):353–7. doi: 10.1038/nature14347. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Turajlic S, Xu H, Litchfield K, Rowan A, Chambers T, Lopez JI, et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. [cited 2024 Feb 1];Cell. 2018 Apr 19;173(3):581–594.:e12. doi: 10.1016/j.cell.2018.03.057. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Spain L, Coulton A, Lobon I, Rowan A, Schnidrig D, Shepherd STC, et al. Late-Stage Metastatic Melanoma Emerges through a Diversity of Evolutionary Pathways. [cited 2024 Feb 1];Cancer Discov. 2023 13(6):1364–85. doi: 10.1158/2159-8290.CD-22-1427. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Foster B, Bagci U, Mansoor A, Xu Z, Mollura DJ. A review on segmentation of positron emission tomography images. [cited 2024 Apr 14];Comput Biol Med. 2014 Jul 1;50:76–96. doi: 10.1016/j.compbiomed.2014.04.014. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bera K, Braman N, Gupta A, Velcheti V, Madabhushi A. Predicting cancer outcomes with radiomics and artificial intelligence in radiology. [cited 2024 Apr 14];Nat Rev Clin Oncol. 2022 Feb 1;19(2):132–46. doi: 10.1038/s41571-021-00560-7. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kaczanowska S, Murty T, Alimadadi A, Contreras CF, Duault C, Subrahmanyam PB, et al. Immune determinants of CAR-T cell expansion in solid tumor patients receiving GD2 CAR-T cell therapy. [cited 2024 Apr 14];Cancer Cell. 2024 Jan 8;42(1):35–51.:e8. doi: 10.1016/j.ccell.2023.11.011. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Gyawali B, Hey SP, Kesselheim AS. Evaluating the evidence behind the surrogate measures included in the FDA’s table of surrogate endpoints as supporting approval of cancer drugs. [cited 2024 Apr 5];EClinicalMedicine. 2020 Apr 1;21 doi: 10.1016/j.eclinm.2020.100332. [Internet] [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES