Next Article in Journal
Inhibition of Sphingosine Kinase 2 Results in PARK2-Mediated Mitophagy and Induces Apoptosis in Multiple Myeloma
Next Article in Special Issue
Developments in Checkpoint Inhibitor Therapy for the Management of Deficient Mismatch Repair (dMMR) Rectal Cancer
Previous Article in Journal
Initial Experiences of Selective RET Inhibitor Selpercatinib in Adults with Metastatic Differentiated Thyroid Carcinoma and Medullary Thyroid Carcinoma: Real-World Case Series in Korea
Previous Article in Special Issue
Mapping Immune Correlates and Surfaceome Genes in BRAF Mutated Colorectal Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Checkpoint Inhibitors-Associated Thrombosis: Incidence, Risk Factors and Management

Department of Medicine, University of Ottawa at The Ottawa Hospital and Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(3), 3032-3046; https://doi.org/10.3390/curroncol30030230
Submission received: 3 February 2023 / Revised: 26 February 2023 / Accepted: 2 March 2023 / Published: 4 March 2023

Abstract

:
Immune checkpoint inhibitors (ICIs) target programmed cell death (PD) 1 receptor and its ligand PD-L1, and have become an integral part of treatment regimens in many cancers including lung cancer, renal cell carcinoma, melanoma, and more. Cancer is associated with a significantly increased risk of venous thromboembolism compared to non-cancer patients, and the risks increase further with anticancer therapies including ICIs. Cancer-associated thrombosis can lead to hospitalizations, delayed cancer treatment, and mortality. While thrombosis was not reported as a major complication in initial clinical trials leading to the approval of ICIs, emerging evidence from post-marketing studies revealed concerning risks of thrombosis in patients receiving ICIs. However, results remained heterogenous given differences in study designs and populations. Recent studies also showed that C-reactive protein dynamics might be an easily accessible biomarker for thrombosis and disease response in this population. In addition, early findings indicated that a commonly used anticoagulant for cancer-associated thrombosis, factor Xa inhibitors, might have potential synergistic antitumor effects when combined with ICIs. Herein we will review the current literature on the incidence, risk factors, and management of thrombosis in patients with cancer receiving ICIs. We aim to provide valuable information for clinicians in managing these patients.

1. Introduction

Tumor cells evade immune destruction by activating immune checkpoint receptor proteins including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) found on T cells, and programmed death ligand 1 (PD-L1) found on tumor cells. A type of novel anticancer therapy termed “immune checkpoint inhibitors (ICIs)” involves monoclonal antibodies that specifically target these proteins and prevent immune escape from tumor cells. Since the approval of the first ICI (ipilimumab) in 2011, seven ICIs have been approved in Canada and the United States in approximately 20 different malignancies (Table 1), including ipilimumab (anti-CTLA-4), nivolumab, pembrolizumab, cemiplimab (anti-PD-1), and atezolizumab, durvalumab, avelumab (anti-PD-L1). As such, patients eligible for ICI treatment significantly increased from 1.5% in 2011 to 43.6% in 2018 [1]. Sustained responses and significant improvement in survival have been seen with these novel therapies, and they have become the mainstay of therapies in many cancers such as melanoma, lung cancer, renal cell carcinoma (RCC), and more [2].
Although thrombosis was not raised as a major concern in initial randomized controlled trials (RCTs) leading to the approval of ICIs, reports of thromboembolic disease had emerged from post-marketing wide use of these agents. Cancer-associated thrombosis is a known phenomenon, and it is estimated that 20% of cancer patients will develop thrombosis during their cancer journey [3]. It can result in hospitalizations, delayed cancer treatment, significant morbidity, and mortality [4,5]. Patients often experience psychosocial and financial distresses from the diagnosis of thrombosis as well [6,7]. Therefore, the optimal prevention and treatment strategies for cancer-associated thrombosis are crucial in the care of cancer patients. Our previous review demonstrated that the reported rate of venous thromboembolism (VTE) was 5-8% in 6 months and over 10% in 12 months in patients receiving ICIs [8]. More studies have been published since then, providing new insights in the incidence, biomarkers, and predictors of thrombosis in this population. Herein we aim to provide an updated narrative review on thrombotic complications in patients with cancer receiving ICIs. For this review, we researched the MEDLINE database from inception to January 31, 2023 by using terms including (“thrombosis” OR “thromboembolism”) AND (“cancer “OR “malignancy”) AND (“immune checkpoint inhibitors” OR “immunotherapy”) and reviewed all relevant prospective or retrospective cohort studies, systematic reviews and meta-analyses, review articles, as well as conference proceedings. We then summarized key information from the literature herein.

2. Mechanism of Thrombosis and Immunotherapy

While the exact mechanisms of ICI-associated thrombosis remain unclear, studies have made strides to understand them. Programmed cell death protein 1 is crucial in downregulating pro-atherogenic T cell responses, so PD-1 antibodies can exacerbate atherosclerotic inflammatory vascular lesions [9] (Figure 1). Studies have shown that ICIs are found to be associated with increased T cell activation and endothelial inflammation, leading to thrombosis formation and accelerated atherosclerosis [10,11]. Similar to other immune-related adverse events associated with ICIs such as colitis and pneumonitis, ICIs release the break on immunoregulatory pathways, lead to an increase in inflammation and related cytokines, activate blood and endothelial cells, as well as release of neutrophil extracellular traps (NETs), and eventually result in thrombosis [12].

3. Incidence of Thrombosis in Patients Receiving Immune Checkpoint Inhibitors

Initial RCTs investigating the efficacy of ICIs as anticancer treatment did not report thrombosis as a major adverse event of concern. An initial meta-analysis of these RCTs showed a modest rate of VTE at 2.7% (95% confidence interval [CI] 1.8–4.0) and an arterial thrombosis (ATE) rate of 1.1% (95% CI 0.5–2.1) [13]. Another meta-analysis of RCTs and prospective studies of ICI use in patients with melanoma and non-small cell lung cancer (NSCLC) also reported similar rates (VTE rates: 1.5% in melanoma and 1.9% in NSCLC) [14]. Most recently, an updated meta-analysis showed no significantly increased risk of VTE (odds ratio [OR] 0.99, 95% CI 0.82–1.19) in patients treated with ICIs compared to non-ICI regimens [15]. However, all of these meta-analyses suffered from the possibility of underreporting of thrombosis events in RCTs in the first place. Solinas et al. excluded 40% of the eligible RCTs from analysis as they did not specifically report rates of thrombosis [13]. In addition, in these studies, thrombosis events were reported as adverse events based on the Common Terminology Criteria for Adverse Events (CTCAE) criteria instead of planned outcomes, and adverse events were often only reported if they were over certain percentages. Significant underreporting of thrombosis events had been shown in previous oncology RCTs with focus on efficacy of cancer therapies compared to thromboprophylaxis trials, which requires close attention for data interpretation [16].
Since the approval and wide clinical use of various ICIs, occurrences of venous and/or arterial thromboses are increasingly reported. Table 2 summarized the results of 27 cohort studies reported to date, with the majority being retrospective cohorts from single centers. Most (80–100%) patients had metastatic cancer, with the most common cancers including NSCLC, melanoma, and RCC. All studies included a variety of different ICIs available in clinical care. The incidence of thrombosis differed widely among studies, due to diverse types of underlying malignancies, concurrent cancer therapies (such as chemotherapy), and variable follow-up durations. In general, the incidence of VTE was 5–8% at 6 months and 10–15% at 12 months (Table 2). Overall, the rates reported in these retrospective cohort studies were higher than those reported from RCTs (1–2% in meta-analyses) [13,14], but not considerably higher when considering the 6-month risks of VTE of 9–10% in ambulatory cancer patients with Khorana score of ≥ 2 receiving chemotherapy [17]. However, given the efficacy of ICIs, the exposure of ICIs can be prolonged, and the risks of thrombosis can continue to accumulate. Sheng et al. showed that the thrombosis rate did not plateau until approximately 30 months in patients with metastatic RCC and 36 months in those with metastatic urothelial cancer [18,19]. Therefore, as patients continue ICIs, the risks of thrombosis can continue to increase. This is worth noting as compared to chemotherapy, for which the highest risk of thrombosis is typically seen within the first 6 months.
In addition to retrospective cohort studies, three Danish population cohort studies reported the incidence of thrombosis in patients on ICIs (Table 3) [43,44,45]. It is interesting to note that the rates of thrombosis appear to be numerically lower in the population studies compared to the above-mentioned retrospective studies, with 6-month VTE rate of 2–4% and 4–7% at 12 months. In these population studies, outcomes were identified by ICD10 codes +/− imaging codes (as compared to individual chart review in retrospective studies), which could account for the difference in event rates. In addition, all three studies were from a single country (Danish registry), as compared to various countries from cohort studies (Table 2).
Whether ICI combinations or combined ICI and chemotherapy would be associated with an increased risk of VTE (compared to single ICI or chemotherapy alone) is also an area of debate. Sussman et al. showed that ICI combinations were associated with a higher risk of VTE compared to single agent ICI [46]. A recent meta-analysis also showed that compared to mono-ICI, combined ICIs were associated with an increased risk of VTE and myocardial infarction in those with NSCLC [14]. However, another study showed that combined ICIs were not associated with higher risks [47]. Similarly, some studies showed an increased risk of ICI–chemotherapy combination compared to chemotherapy alone [31,42], while others showed similar rates of thrombosis in patients receiving ICI alone, chemotherapy alone, or combination [22]. Most recently, in the analysis of an oncology database of 2299 patients with stage IV NSCLC, first-line ICI-based regimens were associated with a 26% reduction in the risk of VTE compared to chemotherapy-based regimens, while the risks were comparable between ICI/chemo versus chemotherapy alone [40]. It is worth noting that a fair comparison of the risks of thrombosis associated with combination of chemotherapy and ICIs to ICI alone or chemotherapy alone could be challenging, as the baseline characteristics of these patients commonly differ significantly.
Data on the incidence or arterial thrombosis were even more scant, with rates within 1-2% over a follow-up period of 6 to 17 months (Table 2). A recent meta-analysis showed an increased risk of arterial thrombosis (OR 1.58, 95% CI 1.21–2.06) associated with ICI regimens [15]. In a matched cohort study of 2842 patients, Drobni et al. revealed that there was a three-fold increased risk of cardiovascular events after the start of ICIs compared to other anticancer therapies. The risks were similarly increased when they compared before and after ICI use in the same patients [25]. They also found ICI to be associated with a > three-fold higher rate of progression of total aortic plaque volume, which can be attenuated with concomitant use of corticosteroids or statins [25].

4. Biomarkers for Thrombosis in Patients on Immune Checkpoint Inhibitors

C-reactive protein (CRP) dynamics was recently identified as a useful biomarker to predict treatment responses associated with ICIs. Significant improvement of objective response, progression-free survival (PFS), and overall survival (OS) had been shown in patients who were CRP flare-responders (CRP levels more than doubled compared to baseline within one month of ICI initiation and subsequently dropped to lower than baseline within 3 months) [48,49]. Therefore, the use of early CRP dynamics as a biomarker in predicting ICI-associated VTE was explored [50]. In a retrospective study of 405 patients receiving ICIs, CRP was measured at baseline within 4 weeks prior to ICI initiation and longitudinally (every 4 weeks) within the first 3 months after ICI initiation. CRP flare was defined as an increase of CRP by a factor of 2.5, while CRP response was defined as a 50% reduction of CRP. In the multivariable analysis accounting death as a competing risk, early CRP flare was associated with a hazard ratio (HR) of VTE of 3.58 (95% CI 1.07–11.94) compared with no CRP flare. Patients with CRP response with no prior flare had the lowest risks of VTE. VTE was independently associated with mortality in those with CRP flare. This study indicated the role of inflammation in the pathophysiology of ICI-associated VTE and revealed a readily available biomarker that has a good potential to predict VTE in this population. Pending further validation, CRP can become a useful tool in this setting.
In another study, blood samples from 25 patients at the time of starting ICIs (15 subsequently developed VTE and 10 did not) were analyzed and showed that patients who subsequently developed VTE had a significant elevation in the numbers of total myeloid-derived suppressor cells (MDSCs) and levels of inflammatory cytokines including ginterleukin-8 (IL-8), IL-1 receptor antagonist, soluble vascular cell adhesion molecule 1 (sVACM-1), and granulocyte–macrophage colony stimulating factor (GM-CSF) [47]. These markers shed light on the mechanism of thrombus formation in patients treated with ICIs and again indicated the importance of immune-mediated inflammation in the process.
As for ICI-associated arterial thrombosis, a small study of 30 patients on ICIs showed that high sensitivity (hs)-troponin T (TnT) ≥ 14 ng/L was associated with a higher risk of cardiovascular outcomes (including cardiovascular death, stroke, transient ischemic attacks, pulmonary embolism, and/or new onset heart failure) [51]. Another retrospective study of 135 patients with metastatic cancer on first-line pembrolizumab revealed that after a mean follow-up of 490 days, hs-troponin I (TnI) > 50 ng/L prior to the first and the second dose of pembrolizumab was an independent predictor of major adverse cardiac events (MACE) (including myocarditis, acute coronary syndrome, heart failure, VTE, cardiovascular hospitalization, and/or mortality) (HR 8.1, 95% CI 1.67–37.4) [52]. hs-TnI > 50 nl/L prior to first dose of pembrolizumab was also associated with increased all-cause mortality. Given these findings, the 2022 European Society of Cardiology (ESC) guidelines on cardio-oncology recommended to monitor cardiac troponin prior to each cycle of ICIs [53].

5. Risk Factors of Thrombosis in Patients on Immune Checkpoint Inhibitors

Understanding risk factors for thrombosis in cancer patients can assist identification of high-risk patients and tailor thromboprophylaxis accordingly. Many risk prediction models had been proposed, among which Khorana score is the most extensively validated. Khorana score was initially derived and validated in 4066 patients enrolled in the Awareness of Neutropenia in Chemotherapy (ANC) Study Group Registry in the United States and had been subsequently validated in many external cohorts including over 30,000 ambulatory cancer patients [54,55]. It includes several easily identified variables in cancer patients undergoing chemotherapy, including the type of malignancy, body mass index ≥ 35 kg/m2, pre-chemotherapy leukocyte count > 11 × 109/L, hemoglobin < 10 g/dL, and platelet count ≥ 350 × 109/L [54]. Recent RCTs confirmed the utility of Khorana score to stratify risks of cancer-associated thrombosis and target thromboprophylaxis in ambulatory cancer patients with Khorana score ≥ 2 (intermediate-high risk) [56,57]. Therefore, although the score has some limitations, it is endorsed by major guidelines to use for risk stratification in ambulatory cancer patients [58,59]. As the development of the Khorana score predated ICIs, whether it can accurately predict thrombosis in patients receiving ICIs remains controversial as studies have shown mixed results. Most studies did not find the Khorana score to carry sufficient predictivity in this population [18,23,29,31,32,33,35,36,37], while a few studies did [27,45,46].
In addition to the Khorana score, many studies attempted to identify additional risk factors for thrombosis in this population. However, risk factors found varied significantly among studies, as most studies were single-center with small sample sizes (Table 4). Potential risk factors identified included (1) patient-related factors, such as female gender [20,28], history of thromboembolism [22,24,27,35,37,38,39,46], younger age [26,27,47], smoking [23,26], and poorer ECOG status [29,39]; (2) cancer-related factors, such as lung cancer [22,25], and metastasis [36,37,47]; and (3) treatment-related factors, such as combined ICI use [46], combined chemotherapy and ICI use, and more. A specific risk prediction model in this population to help identify patients at high risk who could benefit from primary thromboprophylaxis will be of interest.
As for arterial thrombosis, evidence is scant, but few available studies identified risk factors not different from traditional risk factors for arthrosclerosis, such as age, diabetes, hypertension, smoking, history of cardiovascular disease (Table 4).

6. Consequences of Thrombosis in Patients on Immune Checkpoint Inhibitors

Thrombosis can lead to hospitalizations, delay in cancer treatment, and other morbidity and mortality. In a study of 351 patients with metastatic RCC receiving ICIs, 12% (n = 43) developed thrombotic events over a median follow-up of 12.8 months, and 72% of these events resulted in hospitalizations, with 21% having a median delay of 14 days in subsequent ICI doses [18]. In another study of 279 patients with metastatic urothelial cancer, thrombotic events resulted in 83% hospitalization, and 7% delayed ICI treatment with one discontinuation of ICI due to thrombosis [19].
In addition, worsening survival has been shown in patients with thrombosis (either VTE or combined venous and arterial thrombotic events) in this population in many studies [18,19,35,36,37,38,46], while some other (but fewer) studies did not show such an association [26,28,31]. The discrepancy could be due to factors such as patient population, type, and status of cancer.

7. Prevention and Treatment of Thrombosis in Patients on Immune Checkpoint Inhibitors

Recent RCTs including AVERT and CASSINI trials demonstrated the efficacy and safety of prophylactic doses of apixaban and rivaroxaban, respectively, in the prevention of cancer-associated thrombosis in ambulatory cancer patients with intermediate-high risk patients (Khorana score ≥ 2) [56,57]. Major international guidelines have since suggested consideration of primary VTE prophylaxis in this population [58,59]. However, whether this practice can also apply to patients receiving ICIs remains unclear, as Khorana score, derived from a chemotherapy-treated population, had been shown to be suboptimal in risk stratification for patients on ICIs [18,23,29,31,32,33,35,36,37]. Patients with Khorana score of ≥ 2 on chemotherapy are associated with a 6-month risk of VTE of 9–10% [17], and whether the ICI-treated population with a 6-month VTE risk of 5–8% can derive sufficient benefit to warrant primary thromboprophylaxis in all patients receiving ICIs remains investigational. Further identification of high-risk patient population may be needed.
Treatment for cancer-associated VTE in patients receiving ICIs is usually not different from all other cancer-associated thrombosis. Anticoagulation is the mainstay of treatment, with direct oral anticoagulants and low-molecular-weight heparin (LMWH) as the most commonly used anticoagulants in the cancer population; the choice of anticoagulants should take into account patient characteristics, tumor characteristics, risk of bleeding, patient preference, and affordability [59,60].

8. Anticoagulation and Cancer Survival

Preclinical studies showed that coagulation factors such as factor X could assist tumors to escape the immune system, thus contributing to resistance of ICIs. In mice models, factor Xa inhibitors such as rivaroxaban could augment the effects of ICIs against tumor cells and inhibit tumor growth [61,62]. Therefore, whether this can translate into clinical practice—to use factor Xa inhibitors to enhance the antitumor effects of ICIs—is of particular interest. A retrospective study disappointingly showed no difference in response rates, PFS, or OS in patients receiving ICIs and therapeutic anticoagulation compared to those without anticoagulation [63]. However, the anticoagulants used in the study were not limited to factor Xa inhibitors and there was imbalance in baseline characteristics in those who were treated on anticoagulation [63]. In a more recent German study of 280 patients with stage IV melanoma treated with ICIs, again there was no significant difference in the overall response rate, disease control rate, PFS, or OS comparing patients on anticoagulation to those who were not (after adjusting for baseline characteristics differences) [30]. However, when stratified by the type of anticoagulant, the 27 patients who received factor Xa inhibitors had a significant improvement in PFS compared to those on other anticoagulants (median PFS 12 vs. 2 months; HR 0.36, 95% CI 0.2–0.64) and those not on anticoagulants (median PFS 4 months). The OS, as well as overall response rate and disease control rates, were all significantly superior with concomitant factor Xa inhibitors. The improvements in outcomes were not explained by the differences in baseline characteristics. Reassuringly, there were no differences in bleeding events (major and clinically relevant-non major bleeding) among patients receiving factor Xa inhibitors compared to those not on anticoagulation or on different types of anticoagulants during follow-up. The proposed mechanism is that factor Xa inhibitors can penetrate the tumor microenvironment to inhibit signaling function of macrophage-derived factor Xa that promote immune evasion, while LMWH depends on antithrombin for function and is restricted to intravascular space, and thus could not provide synergistic effects to ICIs as factor Xa inhibitors. This is the first study to provide clinical evidence that there could be a synergy between factor Xa inhibitors and ICIs and requires further validation. If confirmed in larger studies, factor Xa inhibitors can play a potential role in optimizing ICI treatment.

9. Conclusions

Immune checkpoint inhibitors are increasingly used and with prolonged duration. Recent studies have shown that rates of VTE in patients receiving ICIs appeared higher than initially reported in RCTs but might be similar or not as high as those receiving chemotherapy. However, the risk can continue to increase with continued ICI use. Arterial thrombosis should also be considered in this population. It is crucial to understand thrombosis as an important adverse event for the optimal management of these patients. The potential synergistic antitumor effects of factor Xa inhibitors and ICI is exciting, but further investigation is needed.

Author Contributions

Conceptualization, T.-F.W. and M.C.; writing—original draft preparation, T.-F.W.; data interpretation: T.-F.W. and M.C.; writing—review and editing, T.-F.W. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

M. Carrier is the recipient of a Tier 1 Research Chair in Cancer and Thrombosis from the Department and Faculty of Medicine at the University of Ottawa.

Conflicts of Interest

T.F.W. reports advisory board honoraria from Servier and Valeo Pharma and grants from Leo Pharma. M.C. reports grants from BMS, Leo Pharma and Pfizer, personal fees from BMS, Leo Pharma, Bayer, Pfizer, Servier, and Sanofi.

References

  1. Haslam, A.; Prasad, V. Estimation of the Percentage of US Patients With Cancer Who Are Eligible for and Respond to Checkpoint Inhibitor Immunotherapy Drugs. JAMA Netw. Open. 2019, 2, e192535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Himmel, M.E.; Saibil, S.D.; Saltman, A.P. Immune checkpoint inhibitors in cancer immunotherapy. CMAJ 2020, 192, E651. [Google Scholar] [CrossRef] [PubMed]
  3. Lyman, G.H.; Eckert, L.; Wang, Y.; Wang, H.; Cohen, A. Venous thromboembolism risk in patients with cancer receiving chemotherapy: A real-world analysis. Oncologists 2013, 18, 1321–1329. [Google Scholar] [CrossRef] [Green Version]
  4. Khorana, A.A.; Francis, C.W.; Culakova, E.; Kuderer, N.M.; Lyman, G.H. Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. J. Thromb. Haemost. 2007, 5, 632–634. [Google Scholar] [CrossRef]
  5. Lloyd, A.J.; Dewilde, S.; Noble, S.; Reimer, E.; Lee, A.Y.Y. What Impact Does Venous Thromboembolism and Bleeding Have on Cancer Patients’ Quality of Life? Value Health 2018, 21, 449–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Noble, S.; Nelson, A.; Scott, J.; Berger, A.; Schmidt, K.; Swarnkar, P.; Lee, A. Patient Experience of Living With Cancer-Associated Thrombosis in Canada (PELICANADA). Res. Pract. Thromb. Haemost. 2020, 4, 154–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Font, C.; Nelson, A.; Garcia-Fernandez, T.; Prout, H.; Gee, P.; Noble, S. Patients’ Experience of Living with Cancer-associated thrombosis in Spain (PELICANOS). Support Care Cancer 2018, 26, 3233–3239. [Google Scholar] [CrossRef]
  8. Wang, T.-F.; Khorana, A.A.; Carrier, M. Thrombotic Complications Associated with Immune Checkpoint Inhibitors. Cancers 2021, 13, 4606. [Google Scholar] [CrossRef]
  9. Jackson, S.P.; Darbousset, R.; Schoenwaelder, S.M. Thromboinflammation: Challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood 2019, 133, 906–918. [Google Scholar] [CrossRef] [Green Version]
  10. Kunimasa, K.; Nishino, K.; Kimura, M.; Inoue, T.; Tamiya, M.; Kumagai, T.; Imamura, F. Pembrolizumab-induced acute thrombosis: A case report. Medicine 2018, 97, e10772. [Google Scholar] [CrossRef] [PubMed]
  11. Cochain, C.; Chaudhari, S.M.; Koch, M.; Wiendl, H.; Eckstein, H.H.; Zernecke, A. Programmed cell death-1 deficiency exacerbates T cell activation and atherogenesis despite expansion of regulatory T cells in atherosclerosis-prone mice. PLoS ONE 2014, 9, e93280. [Google Scholar] [CrossRef] [Green Version]
  12. McCrae, K.R.; Swaidani, S.; Diaz-Montero, C.M.; Khorana, A.A. Old is new again: Emergence of thromboembolic complications in cancer patients on immunotherapy. Thromb. Res. 2022, 213, S51–S57. [Google Scholar] [CrossRef]
  13. Solinas, C.; Saba, L.; Sganzerla, P.; Petrelli, F. Venous and arterial thromboembolic events with immune checkpoint inhibitors: A systematic review. Thromb. Res. 2020, 196, 444–453. [Google Scholar] [CrossRef]
  14. Giustozzi, M.; Becattini, C.; Roila, F.; Agnelli, G.; Mandala, M. Vascular events with immune checkpoint inhibitors in melanoma or non-small cell lung cancer: A systematic review and meta-analysis. Cancer Treat Rev. 2021, 100, 102280. [Google Scholar] [CrossRef]
  15. Ma, Z.; Sun, X.; Zhang, Y.; Li, H.; Sun, D.; An, Z.; Zhang, Y. Risk of Thromboembolic Events in Cancer Patients Treated with Immune Checkpoint Inhibitors: A Meta-analysis of Randomized Controlled Trials. Thromb. Haemost. 2022, 122, 1757–1766. [Google Scholar] [CrossRef] [PubMed]
  16. Chiasakul, T.; Patell, R.; Maraveyas, A.; Carrier, M.; Zwicker, J.I. Discordant reporting of VTE in pancreatic cancer: A systematic review and meta-analysis of thromboprophylaxis versus chemotherapeutic trials. J. Thromb. Haemost. 2021, 19, 489–501. [Google Scholar] [CrossRef] [PubMed]
  17. Ay, C.; Dunkler, D.; Marosi, C.; Chiriac, A.-L.; Vormittag, R.; Simanek, R.; Quehenberger, P.; Zielinski, C.; Pabinger, I. Prediction of venous thromboembolism in cancer patients. Blood 2010, 116, 5377–5382. [Google Scholar] [CrossRef] [PubMed]
  18. Sheng, I.Y.; Gupta, S.; Reddy, C.A.; Angelini, D.; Funchain, P.; Sussman, T.A.; Sleiman, J.; Ornstein, M.C.; McCrae, K.; Khorana, A.A. Thromboembolism in Patients with Metastatic Renal Cell Carcinoma Treated with Immunotherapy. Target Oncol. 2021, 16, 813–821. [Google Scholar] [CrossRef]
  19. Sheng, I.Y.; Gupta, S.; Reddy, C.A.; Angelini, D.; Funchain, P.; Sussman, T.A.; Sleiman, J.; Ornstein, M.C.; McCrae, K.; Khorana, A.A. Thromboembolism in Patients with Metastatic Urothelial Cancer Treated with Immune Checkpoint Inhibitors. Target Oncol. 2022, 17, 563–569. [Google Scholar] [CrossRef]
  20. Hegde, A.M.; Stroud, C.R.; Cherry, C.R.; Yogarajah, M.; Cherukuri, S.D.; Walker, P.R. Incidence and impact of thromboembolic events in lung cancer patients treated with nivolumab. J. Clin. Oncol. 2017, 35 (Suppl. 15), e20624. [Google Scholar] [CrossRef]
  21. Ibrahimi, S.; Machiorlatti, M.; Vesely, S.K.; Malla, M.; Modhia, F.; Jones, S.A.; Cherry, M.A. Incidence of Vascular Thromboembolic Events in Patients Receiving Immunotherapy: A Single Institution Experience. Blood 2017, 130, 4864. [Google Scholar]
  22. Bar, J.; Markel, G.; Gottfried, T.; Percik, R.; Leibowitz-Amit, R.; Berger, R.; Golan, T.; Daher, S.; Taliansky, A.; Dudnik, E.; et al. Acute vascular events as a possibly related adverse event of immunotherapy: A single-institute retrospective study. Eur. J. Cancer 2019, 120, 122–131. [Google Scholar] [CrossRef]
  23. Nichetti, F.; Ligorio, F.; Zattarin, E.; Signorelli, D.; Prelaj, A.; Proto, C.; Galli, G.; Marra, A.; Apollonio, G.; Porcu, L.; et al. Is There an Interplay between Immune Checkpoint Inhibitors, Thromboprophylactic Treatments and Thromboembolic Events? Mechanisms and Impact in Non-Small Cell Lung Cancer Patients. Cancers 2019, 12, 67. [Google Scholar] [CrossRef] [Green Version]
  24. Ando, Y.; Hayashi, T.; Sugimoto, R.; Nishibe, S.; Ito, K.; Kawada, K.; Ikeda, Y.; Yamada, S.; Imaizumi, K. Risk factors for cancer-associated thrombosis in patients undergoing treatment with immune checkpoint inhibitors. Investig. New Drugs 2020, 38, 1200–1206. [Google Scholar] [CrossRef] [Green Version]
  25. Drobni, Z.D.; Alvi, R.M.; Taron, J.; Zafar, A.; Murphy, S.P.; Rambarat, P.K.; Mosarla, R.C.; Lee, C.; Zlotoff, D.A.; Raghu, V.K.; et al. Association Between Immune Checkpoint Inhibitors With Cardiovascular Events and Atherosclerotic Plaque. Circulation 2020, 142, 2299–2311. [Google Scholar] [CrossRef]
  26. Deschênes-Simard, X.; Richard, C.; Galland, L.; Blais, F.; Desilets, A.; Malo, J.; Cvetkovic, L.; Belkaid, W.; Elkrief, A.; Gagné, A.; et al. Venous thrombotic events in patients treated with immune checkpoint inhibitors for non-small cell lung cancer: A retrospective multicentric cohort study. Thromb. Res. 2021, 205, 29–39. [Google Scholar] [CrossRef]
  27. Gong, J.; Drobni, Z.D.; Alvi, R.M.; Murphy, S.P.; Sullivan, R.J.; Hartmann, S.E.; Gilman, H.K.; Lee, H.; Zubiri, L.; Raghu, V.K.; et al. Immune checkpoint inhibitors for cancer and venous thromboembolic events. Eur. J. Cancer 2021, 158, 99–110. [Google Scholar] [CrossRef] [PubMed]
  28. Gutierrez-Sainz, L.; Martinez-Marin, V.; Viñal, D.; Martinez-Perez, D.; Pedregosa, J.; Garcia-Cuesta, J.A.; Feliu, J. Incidence of venous thromboembolic events in cancer patients receiving immunotherapy: A single-institution experience. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. Natl. Cancer Inst. Mex. 2021, 23, 1245–1252. [Google Scholar] [CrossRef]
  29. Guven, D.C.; Aksun, M.S.; Sahin, T.K.; Aktepe, O.H.; Yildirim, H.C.; Taban, H.; Ceylan, F.; Kertmen, N.; Arik, Z.; Dizdar, O.; et al. Poorer baseline performance status is associated with increased thromboembolism risk in metastatic cancer patients treated with immunotherapy. Support. Care Cancer Off. J. Multinatl. Assoc. Support. Care Cancer 2021, 29, 5417–5423. [Google Scholar] [CrossRef] [PubMed]
  30. Haist, M.; Stege, H.; Pemler, S.; Heinz, J.; Fleischer, M.I.; Graf, C.; Ruf, W.; Loquai, C.; Grabbe, S. Anticoagulation with Factor Xa Inhibitors Is Associated with Improved Overall Response and Progression-Free Survival in Patients with Metastatic Malignant Melanoma Receiving Immune Checkpoint Inhibitors-A Retrospective, Real-World Cohort Study. Cancers 2021, 13, 5103. [Google Scholar] [CrossRef] [PubMed]
  31. Hill, H.; Robinson, M.; Lu, L.; Slaughter, D.; Amin, A.; Mileham, K.; Patel, J.N. Venous thromboembolism incidence and risk factors in non-small cell lung cancer patients receiving first-line systemic therapy. Thromb. Res. 2021, 208, 71–78. [Google Scholar] [CrossRef] [PubMed]
  32. Icht, O.; Darzi, N.; Shimony, S.; Jacobi, O.; Reinhorn, D.; Landman, Y.; Leader, A. Venous thromboembolism incidence and risk assessment in lung cancer patients treated with immune checkpoint inhibitors. J. Thromb. Haemost. 2021, 19, 1250–1258. [Google Scholar] [CrossRef] [PubMed]
  33. Kewan, T.; Ko, T.; Flores, M.; Sallam, Y.; Haddad, A.; Daw, H. Prognostic impact and risk factors of cancer-associated thrombosis events in stage-IV cancer patients treated with immune checkpoint inhibitors. Eur. J. Haematol. 2021, 106, 682–688. [Google Scholar] [CrossRef]
  34. Madison, C.J.; Melson, R.A.; Conlin, M.J.; Gundle, K.R.; Thompson, R.F.; Calverley, D.C. Thromboembolic risk in patients with lung cancer receiving systemic therapy. Br. J. Haematol. 2021, 194, 179–190. [Google Scholar] [CrossRef]
  35. Moik, F.; Chan, W.-S.E.; Wiedemann, S.; Hoeller, C.; Tuchmann, F.; Aretin, M.-B.; Fuereder, T.; Zöchbauer-Müller, S.; Preusser, M.; Pabinger, I.; et al. Incidence, risk factors, and outcomes of venous and arterial thromboembolism in immune checkpoint inhibitor therapy. Blood 2021, 137, 1669–1678. [Google Scholar] [CrossRef]
  36. Alma, S.; Eloi, D.; Léa, V.; Julie, C.; Valérie, M.; Pierre, G.; Hilgers, W.; Philippe, G.; Christine, Z.; Philippe, D. Incidence of venous thromboembolism and discriminating capacity of Khorana score in lung cancer patients treated with immune checkpoint inhibitors. J. Thromb. Thrombolysis 2022, 54, 287–294. [Google Scholar] [CrossRef]
  37. Bjørnhart, B.; Kristiansen, C.; Asmussen, J.; Hansen, K.H.; Wedervang, K.; Jørgensen, T.L.; Herrstedt, J.; Schytte, T. Clinical impact of venous thromboembolism in non-small cell lung cancer patients receiving immunotherapy. Thromb. Res. 2023, 221, 164–172. [Google Scholar] [CrossRef]
  38. Cánovas, M.S.; Garay, D.F.; Moran, L.O.; Pérez, J.R.; Rubio, C.M.G.; de Mena, M.L.; Portero, B.O.; Castro, J.B.; Lage, Y.; Lavin, D.C.; et al. Immune checkpoint inhibitors-associated thrombosis in patients with lung cancer and melanoma: A study of the Spanish society of medical oncology (SEOM) thrombosis and cancer group. Clin. Transl. Oncol. 2022, 24, 2010–2020. [Google Scholar] [CrossRef]
  39. Endo, S.; Honda, T.; Kawahara, T.; Sakakibara, R.; Mitsumura, T.; Okamoto, T.; Miyazaki, Y. Profile of metastatic lung cancer patients susceptible to development of thromboembolism during immunotherapy. Cancer Treat. Res. Commun. 2022, 31, 100547. [Google Scholar] [CrossRef] [PubMed]
  40. Khorana, A.; Palaia, J.; Rosenblatt, L.; Pisupati, R.; Huang, N.; Nguyen, C.; Barron, J.; Gallagher, K.; Bond, T.C. Venous thromboembolism incidence and risk factors associated with immune checkpoint inhibitors among patients with advanced non-small cell lung cancer. J. Immunother. Cancer 2023, 11, e006072. [Google Scholar] [CrossRef] [PubMed]
  41. May, S.B.; La, J.; Milner, E.; Riaz, N.; Amos, C.; Garcia, D.A.; Carrier, M.; Zakai, N.; Do, N.V.; Brophy, M.; et al. Venous Thromboembolism Risk in Cancer Patients Receiving First-Line Immune Checkpoint Inhibitor Vs. Chemotherapy. Blood 2022, 140, 7968–7970. [Google Scholar] [CrossRef]
  42. Sanfilippo, K.M.; Luo, S.; Lyman, G.H.; Calverley, D.C.; Kuderer, N. Identification of Risk Factors for and Development of a Predictive Model for Immunotherapy-Associated Venous Thromboembolism (VTE) in Patients with Non-Small Cell Lung Cancer. Blood 2022, 140, 2803–2804. [Google Scholar] [CrossRef]
  43. Mulder, F.I.; Horváth-Puhó, E.; van Es, N.; van Laarhoven, H.W.M.; Pedersen, L.; Moik, F.; Ay, C.; Büller, H.R.; Sørensen, H.T. Venous thromboembolism in cancer patients: A population-based cohort study. Blood 2021, 137, 1959–1969. [Google Scholar] [CrossRef] [PubMed]
  44. Moik, F.; Ay, C.; Horváth-Puhó, E.; Pabinger, I.; Mulder, F.I.; van Es, N.; Sørensen, H.T. Risk of Venous and Arterial Thromboembolic Events in Patients Receiving Targeted Anti-cancer Therapy–A Nationwide Cohort Study. Available online: https://abstracts.isth.org/abstract/risk-of-venous-and-arterial-thromboembolic-events-in-patients-receiving-targeted-anti-cancer-therapy-a-nationwide-cohort-study/ (accessed on 2 February 2023).
  45. Overvad, T.F.; Skjøth, F.; Piazza, G.; Noble, S.; Ording, A.G.; Larsen, T.B.; Nielsen, P.B. The Khorana score and venous and arterial thrombosis in patients with cancer treated with immune checkpoint inhibitors: A Danish cohort study. J. Thromb. Haemost. JTH 2022, 20, 2921–2929. [Google Scholar] [CrossRef]
  46. Sussman, T.A.; Li, H.; Hobbs, B.; Funchain, P.; McCrae, K.R.; Khorana, A.A. Incidence of thromboembolism in patients with melanoma on immune checkpoint inhibitor therapy and its adverse association with survival. J. Immunother. Cancer 2021, 9, e001719. [Google Scholar] [CrossRef] [PubMed]
  47. Roopkumar, J.; Swaidani, S.; Kim, A.S.; Thapa, B.; Gervaso, L.; Hobbs, B.P.; Wei, W.; Alban, T.J.; Funchain, P.; Kundu, S.; et al. Increased Incidence of Venous Thromboembolism with Cancer Immunotherapy. Medicine 2021, 2, 423–434. [Google Scholar] [CrossRef] [PubMed]
  48. Fukuda, S.; Saito, K.; Yasuda, Y.; Kijima, T.; Yoshida, S.; Yokoyama, M.; Ishioka, J.; Matsuoka, Y.; Kageyama, Y.; Fujii, Y. Impact of C-reactive protein flare-response on oncological outcomes in patients with metastatic renal cell carcinoma treated with nivolumab. J. Immunother. Cancer 2021, 9, e001564. [Google Scholar] [CrossRef] [PubMed]
  49. Klümper, N.; Saal, J.; Berner, F.; Lichtensteiger, C.; Wyss, N.; Heine, A.; Bauernfeind, F.G.; Ellinger, J.; Brossart, P.; Diem, S.; et al. C reactive protein flare predicts response to checkpoint inhibitor treatment in non-small cell lung cancer. J. Immunother Cancer 2022, 10, e004024. [Google Scholar] [CrossRef]
  50. Moik, F.; Riedl, J.; Barth, D.; Chan, W.-S.E.; Wiedemann, S.; Höller, C.; Fuereder, T.; Jost, P.; Pabinger, I.; Preusser, M.; et al. Early Dynamics of C-Reactive Protein Predict Risk of Venous Thromboembolism in Patients with Cancer Treated with Immune Checkpoint Inhibitors. Blood 2022, 140, 1250–1251. [Google Scholar] [CrossRef]
  51. Petricciuolo, S.; Delle Donne, M.G.; Aimo, A.; Chella, A.; De Caterina, R. Pre-treatment high-sensitivity troponin T for the short-term prediction of cardiac outcomes in patients on immune checkpoint inhibitors. Eur. J. Clin. Investig. 2021, 51, e13400. [Google Scholar] [CrossRef]
  52. Waissengein, B.; Abu Ata, B.; Merimsky, O.; Shamai, S.; Wolf, I.; Arnold, J.H.; Bar-On, T.; Banai, S.; Khoury, S.; Laufer-Perl, M. The predictive value of high sensitivity troponin measurements in patients treated with immune checkpoint inhibitors. Clin. Res. Cardiol. Off. J. Ger. Card. Soc. 2022. ahead of print. [Google Scholar] [CrossRef] [PubMed]
  53. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Zamorano, J.L. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef] [PubMed]
  54. Khorana, A.A.; Kuderer, N.M.; Culakova, E.; Lyman, G.H.; Francis, C.W. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 2008, 111, 4902–4907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Mulder, F.I.; Candeloro, M.; Kamphuisen, P.; Di Nisio, M.; Bossuyt, P.M.; Guman, N.; Smit, K.; Büller, H.R.; Van Es, N. The Khorana score for prediction of venous thromboembolism in cancer patients: A systematic review and meta-analysis. Haematologica 2019, 104, 1277–1287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Carrier, M.; Abou-Nassar, K.; Mallick, R.; Tagalakis, V.; Shivakumar, S.; Schattner, A.; Kuruvilla, P.; Hill, D.; Spadafora, S.; Marquis, K.; et al. Apixaban to Prevent Venous Thromboembolism in Patients with Cancer. N. Engl. J. Med. 2019, 380, 711–719. [Google Scholar] [CrossRef] [PubMed]
  57. Khorana, A.A.; Soff, G.A.; Kakkar, A.K.; Vadhan-Raj, S.; Riess, H.; Wun, T.; Streiff, M.B.; Garcia, D.A.; Liebman, H.A.; Belani, C.P.; et al. Rivaroxaban for Thromboprophylaxis in High-Risk Ambulatory Patients with Cancer. N. Engl. J. Med. 2019, 380, 720–728. [Google Scholar] [CrossRef] [PubMed]
  58. Key, N.S.; Khorana, A.A.; Kuderer, N.M.; Bohlke, K.; Lee, A.Y.; Arcelus, J.I.; Wong, S.L.; Balaban, E.P.; Flowers, C.R.; Francis, C.W.; et al. Venous Thromboembolism Prophylaxis and Treatment in Patients With Cancer: ASCO Clinical Practice Guideline Update. J. Clin. Oncol. 2020, 38, 496–520. [Google Scholar] [CrossRef]
  59. Lyman, G.H.; Carrier, M.; Ay, C.; Di Nisio, M.; Hicks, L.K.; Khorana, A.A.; Leavitt, A.D.; Lee, A.Y.Y.; Macbeth, F.; Morgan, R.L.; et al. American Society of Hematology 2021 guidelines for management of venous thromboembolism: Prevention and treatment in patients with cancer. Blood Adv. 2021, 5, 927–974. [Google Scholar] [CrossRef]
  60. Carrier, M.; Blais, N.; Crowther, M.; Kavan, P.; Le Gal, G.; Moodley, O.; Shivakumar, S.; Suryanarayan, D.; Tagalakis, V.; Wu, C.; et al. Treatment Algorithm in Cancer-Associated Thrombosis: Updated Canadian Expert Consensus. Curr. Oncol. 2021, 28, 5434–5451. [Google Scholar] [CrossRef]
  61. Graf, C.; Wilgenbus, P.; Pagel, S.; Pott, J.; Marini, F.; Reyda, S.; Kitano, M.; Macher-Göppinger, S.; Weiler, H.; Ruf, W. Myeloid cell-synthesized coagulation factor X dampens antitumor immunity. Sci. Immunol. 2019, 4, eaaw8405. [Google Scholar] [CrossRef]
  62. Ruf, W.; Graf, C. Coagulation signaling and cancer immunotherapy. Thromb. Res. 2020, 191 (Suppl. 1), S106–S111. [Google Scholar] [CrossRef] [PubMed]
  63. Johannet, P.; Sawyers, A.; Gulati, N.; Donnelly, D.; Kozloff, S.; Qian, Y.; Floristan, A.; Hernando, E.; Zhong, J.; Osman, I. Treatment with therapeutic anticoagulation is not associated with immunotherapy response in advanced cancer patients. J. Transl. Med. 2021, 19, 47. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Summary of mechanism, incidence, consequences, prevention, and treatment of immune checkpoint inhibitor-associated thrombosis. Figure was created with BioRender.com and adapted from “Immune Checkpoint Inhibitor Against Tumor Cell”, by BioRender.com (2023). Retrieved from https://app.biorender.com/biorender-templates (accessed on 2 February 2023).
Figure 1. Summary of mechanism, incidence, consequences, prevention, and treatment of immune checkpoint inhibitor-associated thrombosis. Figure was created with BioRender.com and adapted from “Immune Checkpoint Inhibitor Against Tumor Cell”, by BioRender.com (2023). Retrieved from https://app.biorender.com/biorender-templates (accessed on 2 February 2023).
Curroncol 30 00230 g001
Table 1. Summary of approved immune checkpoint inhibitors (and their indications).
Table 1. Summary of approved immune checkpoint inhibitors (and their indications).
Immune Checkpoint InhibitorsTargetApproved Indication
IpilimumabCTLA-4Melanoma
NSCLC
RCC
Colorectal cancer
Malignant pleural mesothelioma
PembrolizumabPD-1Melanoma
NSCLC
Urothelial carcinoma
RCC
Bladder cancer
Esophageal/esophagogastric junction cancer
Colorectal cancer
Endometrial cancer
Cervical cancer
Breast cancer
Head and neck squamous cell carcinoma
Hodgkin lymphoma
Primary mediastinal B cell lymphoma
NivolumabPD-1Melanoma
NSCLC
RCC
Head and neck squamous cell carcinoma
Classical Hodgkin lymphoma
Hepatocellular carcinoma
CemiplimabPD-1NSCLC
Cutaneous squamous cell carcinoma
Cutaneous basal cell carcinoma
Cervical cancer
AtezolizumabPD-L1NSCLC
Small cell lung cancer
Urothelial carcinoma
AvelumabPD-L1Urothelial carcinoma
Merkel cell carcinoma
DurvalumabPD-L1NSCLC
Urothelial carcinoma
Mostly approved for unresected or metastatic cancers, refer to individual monographs for detailed approval indications for each cancer. Abbreviations: NSCLC: non-small cell lung cancer; RCC: renal cell carcinoma.
Table 2. Summary of incidence rates of venous and arterial thrombosis from retrospective studies of cancer patients receiving immune checkpoint inhibitors (studies missing follow-up durations were not included).
Table 2. Summary of incidence rates of venous and arterial thrombosis from retrospective studies of cancer patients receiving immune checkpoint inhibitors (studies missing follow-up durations were not included).
StudyCountry NType of Cancer Stage IVFollow-up [Median (IQR)] VTE
Incidence % (95% CI)
ATE
Incidence % (95% CI)
Hegde et al. 2017 [20] AbstractUSA76LungN/A10.8 mo18.42.6
Ibrahimi et al. 2017 [21]
Abstract
USA154Lung 20.8% Melanoma 20.1%
Ovarian 12.3%
92%7 mo (198 days)10.40
Bar et al. 2019 [22]Israel1215All cancers
Melanoma 40.5% Lung 28.7%
N/A12 moAVE (MI, stroke, PE, DVT)
6 mo: 4.9
12 mo: 5.8
Nichetti et al. 2019 [23] Italy217NSCLC95.4% 37.8 mo7.46.5
Ando et al. 2020 [24] Japan122Lung, kidney, stomach, urothelial, melanomaN/AN/A
Time to thrombosis 90 days (range 6–178)
4.14.9
Drobni et al. 2020 [25]USA2842All cancers
NSCLC 28.8% Melanoma 27.9%
N/A2 yearsN/AComposite: 5.35/100 person-yrs
MI: 2.49
Stroke: 2.08
Deschênes-Simard et al. 2021 [26]Canada593NSCLC87.2% 12.7 (4.9–22.7) mo9.9 (7.5–12.3)
76.5 (59.9–97.8) per 1000 person-years
1.3
Gong et al. 2021 [27]USA2854All cancers
NSCLC 28.4% Melanoma 28.2%
N/A194 days (IQR 65–412)6 mo: 7.4
12 mo: 13.8
N/A
Gutierrez-Sainz et al. 2021 [28]Spain229Lung 48%
Melanoma 23.6%
RCC 11.8%
96.5%9.8 mo7 (4–10)N/A
Guven et al. 2021 [29]Turkey133RCC 26.3%
Melanoma 24.1% NSCLC 18.8%
100%10.1 (5.8–18.5) mo11.3N/A
Haist et al. 2021 [30]Germany280Melanoma 100%28 mo (95% CI 23.4–32.6)12.54.3
Hill et al. 2021 [31]USA435
(a) ICI: 171
(b) ICI+chemo: 157
(c) chemo then durvalumab: 107
NSCLC47%N/A6 mo:
(a) 7.6 (4.3–12.2)
(b) 9.9 (5.8–15.3)
(c) 9.4 (4.8–15.8)
12 mo:
(a) 9.0 (5.3–14.0)
(b) 12.8 (7.8–19.0)
(c) 12.2 (6.8–19.2)
N/A
Icht et al. 2021 [32]Israel176NSCLC85.8%6 mo (187 days)4.5 (2.1–8.3)N/A
Kewan et al. 2021 [33]USA552All cancers
NSCLC 47.3%
100%12.1 mo12.11.3
Madison et al. 2021 [34] ^USA6127LungN/A6 mo6.32.6
Moik et al. 2021 [35]Austria672Melanoma 30.4%
NSCLC 24.1%
RCC 11%
85.8%8.5 mo6 mo: 5.0 (3.4–6.9)
12mo: 7.0 (5.1–9.3)
Overall: 12.9 (8.2–18.5)
6 mo: 1.0 (0.4–2.0)
12 mo: 1.8 (0.7–3.6)
Overall 1.8 (0.7–3.6)
Roopkumar et al. 2021USA1686Lung 49.6% Melanoma 13.2%90.3%438 days (range 7–1971)6 mo: 7.1
12 mo: 10.9
Overall: 24
N/A
Sheng et al. 2021 [18]USA351RCC100%12.8 mo112
Total thromboembolism:
6 mo: 4.4 (2.6–6.9)
12 mo: 9.8 (6.8–13.4)
Sussman et al. 2021 USA228Melanoma81.1%27.3 mo6 mo: 8.0 (4.9–12.0)
12 mo: 12.9 (8.9–17.7)
6 mo: 2.2 (0.8–4.8)
12 mo: 4.5 (2.3–7.8)
Alma et al. 2022 [36] France481Lung 86%9.8 mo9.8N/A
Bjornhart et al. 2022 [37]Denmark146 prospective (A) *NSCLC87%16.5 mo6 mo: 13.0
12 mo: 14.4
Overall: 14
N/A
426 retrospective (B)6 mo: 4.9
12 mo: 5.6
Overall: 6
Canovas et al. 2022 [38]Spain665Lung91.2%14 mo6.91.5
All thrombosis: 8.4 (6.23–10.6)
291Melanoma82.5%17 mo4.81.0
All thrombosis: 5.8 (3.34–9.18)
Endo et al. 2022 [39]Japan120Lung62.5%within 6 mo2.54.2
Khorana et al. 2023 [40] ^USA(a) ICI: 605
(b) ICI+chemo: 602
NSCLC100%9.1 mo6 mo:
(a) 8.1
(b) 12.8
12 mo:
(a) 13.5 (10.6–16.5)
(b) 22.4 (20.2–24.5)
N/A
May et al. 2022 abstract [41] ^ USA1823All cancersN/A6 mo7.3N/A
Sanfilippo et al. 2022 abstract [42]^ USA1754All cancers77.9%6 mo4.1N/A
Sheng et al. 2022 [19] USA279Urothelial 100%5.6 mo132
Total thromboembolism:
6 mo: 9.1 (6.0–13.0)
12 mo: 13.6 (9.6–18.4)
* A prospective cohort employed screening-detected VTE by computed tomography venography and pulmonary angiography. ^ Outcomes identified by ICD codes. Gong: VTE after ICI compared to pre, HR 4.98 (3.65–8.59). Abbreviations: ATE—arterial thrombosis; AVE—arterial event; CI—confidence interval; DVT—deep vein thrombosis; ICI—immune checkpoint inhibitor; MI—myocardial infarction; mo—months; N/A—not available; NSCLC—non-small cell lung cancer; PE—pulmonary embolism; RCC—renal cell carcinoma; USA—United States of America; VTE—venous thromboembolism.
Table 3. Summary of incidence of VTE and ATE from population cohort studies (all from Danish registries).
Table 3. Summary of incidence of VTE and ATE from population cohort studies (all from Danish registries).
StudyNType of cancerFollow-up (mo)VTE (%), (95% CI)ATE (%), (95% CI)
Mulder et al. 2021 [44]370All cancers64.1 (2.3–6.7)N/A
127.1 (4.2–11.1)
Moik et al. 2021 [44] Abstract3259All cancers63.9 (3.3–4.7)1.3 (0.9–1.8)
125.7 (4.9–6.6)2.2 (1.7–2.8)
247.3 (6.2–8.4)3.1 (2.4–3.8)
Overvad et al. 2022 [45]3946All cancers62.61.3
123.81.9
Abbreviations: ATE—arterial thrombosis; CI—confidence interval; mo—months; VTE—venous thromboembolism.
Table 4. Risk factors for thrombosis and mortality identified in patient cohorts receiving immune checkpoint inhibitors.
Table 4. Risk factors for thrombosis and mortality identified in patient cohorts receiving immune checkpoint inhibitors.
Study Risk factors for Thrombosis (Multivariable)Risk Factors for Mortality
Hegde et al. 2017 [20] (Abstract)Female VTE before ICI
Bar et al. 2019 [22]NSCLC
H/o AVE
Hypertension
Dyslipidemia
AVE
Nichetti et al. 2019 [23]Current smoker
PD-L1 > 50%
TE
Ando et al. 2020 [24]h/o thromboembolismN/A
Drobni et al. 2020 [25]Overall study:
ICIs, age, h/o stroke, diabetes, hypertension, NSCLC, male, h/o radiation
N/A
Deschênes-Simard et al. 2021 [26]Age < 65
Higher PD-L1 level
Smoking
<12 mo from diagnosis to ICIs
VTE is not correlated with survival
Gong et al. 2021 [27]Age ≤ 65
Khorana score ≥ 2
h/o hypertension
Strong trend: h/o VTE (HR 1.42, 95% CI 0.99–2.06)
(melanoma is associated with decreased risks)
N/A
Gutierrez-Sainz et al. 2021 [28]Female
Melanoma
VTE is not an independent risk factor
Guven et al. 2021 [29]ECOG ≥ 1VTE (trend, not significant)
Hill et al. 2021 [31]Cancer treatment types (ICI-chemotherapy, targeted therapies)
Smoking
VTE was not associated with significantly worsened survival
Icht et al. 2021 [32]N/AVTE
Kewan et al. 2021 [33]Anticoagulation at the time of ICI (univariate)Khorana score
Moik et al. 2021 [35]h/o VTEVTE after ICI
Roopkumar et al. 2021 [47]Younger age
Metastasis
Biomarkers
VTE
Sheng et al. 2021 [18]NoneThromboembolism, IMDC score, and/or Khorana score
Sussman et al. 2021 [46]Combination ICI
Khorana score ≥ 1
h/o CAD
Anticoagulation at treatment start
VTE
Alma et al. 2022 [36]Metastasis
BMI
VTE
Bjornhart et al. 2022 [37]h/o VTE
ICI as first-line treatment
Other mets (non-brain, liver, bone)
VTE
Canovas et al. 2022 [38]
Lung cancer cohort
Hgb < 10.9 g/dL at the start of ICI
NLR < 4.55
h/o thrombosis
Thrombosis
Canovas et al. 2022 [38]
Melanoma cohort
LDH > 198 U/L
NLR > 3.01
Thrombosis
Endo et al. 2022 [39]ECOG ≥ 2 and history of thromboembolismN/A
Khorana et al. 2023 [40]History of radiation
Body mass index ≥ 40 kg/m2
N/A
Sanfilippo et al. 2022 [42] (Abstract) ICI-chemotherapy (vs ICI alone)
Severe frailty by CA frailty index
N/A
Sheng et al. 2022 [19]NoneThromboembolism, Bajorin score 1 and 2
Abbreviations: ATE—arterial thrombosis; AVE—acute vascular events; CAD—coronary artery disease; DVT—deep vein thrombosis; ECOG—Eastern Cooperative Oncology Group; Hgb—hemoglobin; h/o—history of; ICIs—immune checkpoint inhibitors; IMDC: International Metastatic Renal Cell Carcinoma Database Consortium; IQR—interquartile range; KS—Khorana score; MI—myocardial infraction; LDH—lactate dehydrogenase; mo—months; N/A—not available; NLR—neutrophil/lymphocyte ratio; NSCLC—non-small cell lung cancer; PE—pulmonary embolism; TE—thromboembolism; VTE—venous thromboembolism.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, T.-F.; Carrier, M. Immune Checkpoint Inhibitors-Associated Thrombosis: Incidence, Risk Factors and Management. Curr. Oncol. 2023, 30, 3032-3046. https://doi.org/10.3390/curroncol30030230

AMA Style

Wang T-F, Carrier M. Immune Checkpoint Inhibitors-Associated Thrombosis: Incidence, Risk Factors and Management. Current Oncology. 2023; 30(3):3032-3046. https://doi.org/10.3390/curroncol30030230

Chicago/Turabian Style

Wang, Tzu-Fei, and Marc Carrier. 2023. "Immune Checkpoint Inhibitors-Associated Thrombosis: Incidence, Risk Factors and Management" Current Oncology 30, no. 3: 3032-3046. https://doi.org/10.3390/curroncol30030230

Article Metrics

Back to TopTop