We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Successful treatment with carboplatin and paclitaxel in melanoma progression after immune-related adverse events

    Özgecan Dülgar‡

    Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA

    ,
    Aditi Saha‡

    Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA

    ,
    Kelly M Elleson

    Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA

    &
    Joseph Markowitz

    *Author for correspondence: Tel.: +1 813 745 8581;

    E-mail Address: joseph.markowitz@moffitt.org

    Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA

    Department of Oncologic Sciences, Morsani School of Medicine University of South Florida, Tampa, FL 33612, USA

    Published Online:https://doi.org/10.2217/imt-2022-0213

    The overall survival of melanoma patients has improved using antibodies targeting immune checkpoints (anti-PD-1, anti-CTLA-4 and anti-LAG-3). Systemic chemotherapy was administered in melanoma for many years with limited effectiveness. Here we report a case of a patient who experienced immune-mediated adverse effects from checkpoint blockade therapy and subsequently responded to chemotherapy. The patient presented with melanoma and paraneoplastic digital ischemia. She received a combination of ipilimumab/nivolumab and experienced G3 myocarditis, followed by melanoma progression after a steroid taper. This patient achieved a partial and durable response with platinum and taxane-based chemotherapy. This report suggests the possibility of a subset of patients who experience progression after immune-based side effects where chemotherapy may be effective in the modern age of melanoma treatment.

    Plain language summary

    We describe a patient with a type of skin cancer called melanoma. At first, we tried to treat it with a medication that made the patient's body's defense system fight against it, but it caused problems with her heart so we had to stop it. Instead, we used another type of treatment called chemotherapy, which worked. The patient remains off therapy 4.5 years later. The lesson learned from this case is that chemotherapy is still helpful in certain situations. The initial treatment did not stop the melanoma growth. In addition, the patient had life-threatening toxicity.

    Tweetable abstract

    Chemotherapy may be useful for melanoma patients intolerant to checkpoint blockade therapy. Written informed consent was obtained from the patient to publish a case report.

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Wolchok JD, Chiarion-Sileni V, Gonzalez R et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 377(14), 1345–1356 (2017).
    • 2. Larkin J, Chiarion-Sileni V, Gonzalez R et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381(16), 1535–1546 (2019).
    • 3. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N. Engl. J. Med. 378(2), 158–168 (2018).
    • 4. Wang Y, Zhou S, Yang F et al. Treatment-related adverse events of PD-1 and PD-L1 inhibitors in clinical trials: a systematic review and meta-analysis. JAMA Oncol. 5(7), 1008–1019 (2019).
    • 5. Johnson DB, Balko JM, Compton ML et al. Fulminant myocarditis with combination immune checkpoint blockade. N. Engl. J. Med. 375(18), 1749–1755 (2016). • Describes myocarditis in the melanoma population treated with combination checkpoint blockade.
    • 6. Matzen E, Bartels LE, Logstrup B, Horskaer S, Stilling C, Donskov F. Immune checkpoint inhibitor-induced myocarditis in cancer patients: a case report and review of reported cases. Cardiooncology 7(1), 27 (2021).
    • 7. Jiménez-Alejandre R, Ruiz-Fernández I, Martín P. Pathophysiology of immune checkpoint inhibitor-induced myocarditis. Cancers (Basel) 14(18), (2022).
    • 8. Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 11(2), 141–151 (1999).
    • 9. Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc. Natl Acad. Sci. USA 102(33), 11823–11828 (2005).
    • 10. Nishimura H, Okazaki T, Tanaka Y et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291(5502), 319–322 (2001).
    • 11. Okazaki T, Tanaka Y, Nishio R et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat. Med. 9(12), 1477–1483 (2003).
    • 12. Serna-Higuita LM, Amaral T, Forschner A et al. Association between immune-related adverse events and survival in 319 stage IV melanoma patients treated with PD-1-based immunotherapy: an approach based on clinical chemistry. Cancers (Basel) 13(23), (2021).
    • 13. Thompson JA, Schneider BJ, Brahmer J et al. NCCN Guidelines Insights: Management of Immunotherapy-Related Toxicities, Version 1.2020. J. Natl Compr. Canc. Netw. 18(3), 230–241 (2020). • Describes how to treat immune-related adverse events.
    • 14. Gambichler T, Strutzmann S, Tannapfel A, Susok L. Paraneoplastic acral vascular syndrome in a patient with metastatic melanoma under immune checkpoint blockade. BMC Cancer 17(1), 327 (2017).
    • 15. Zenati N, Charles J, Templier I, Blaise S. Digital ischaemia with fingertip ulcers during ipilimumab therapy. Ann. Dermatol. Venereol. 147(3), 212–216 (2020).
    • 16. Poszepczynska-Guigne E, Viguier M, Chosidow O, Orcel B, Emmerich J, Dubertret L. Paraneoplastic acral vascular syndrome: epidemiologic features, clinical manifestations, and disease sequelae. J. Am. Acad. Dermatol. 47(1), 47–52 (2002).
    • 17. Jud P, Raggam RB, Hafner F. Paraneoplastic acral vascular syndrome. CMAJ 192(46), E1470 (2020).
    • 18. Falanga A, Marchetti M, Vignoli A. Coagulation and cancer: biological and clinical aspects. J. Thromb. Haemostas. 11(2), 223–233 (2013).
    • 19. Makunts T, Saunders IM, Cohen IV et al. Myocarditis occurrence with cancer immunotherapy across indications in clinical trial and post-marketing data. Sci. Rep. 11(1), 17324 (2021).
    • 20. Palaskas N, Lopez-Mattei J, Durand JB, Iliescu C, Deswal A. Immune checkpoint inhibitor myocarditis: pathophysiological characteristics, diagnosis, and treatment. J. Am. Heart Assoc. 9(2), e013757 (2020).
    • 21. Salem JE, Manouchehri A, Moey M et al. Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol. 19(12), 1579–1589 (2018).
    • 22. Maeda T, Yoshino K, Nagai K et al. The efficacy of platinum-based chemotherapy for immune checkpoint inhibitor-resistant advanced melanoma. Acta Oncol. 58(3), 379–381 (2019). • Describes how to treat immune-related adverse events.
    • 23. Goldinger SM, Buder-Bakhaya K, Lo SN et al. Chemotherapy after immune checkpoint inhibitor failure in metastatic melanoma: a retrospective multicentre analysis. Eur. J. Cancer 162, 22–33 (2022).
    • 24. Gaughan EM, Horton BJ. Outcomes from cytotoxic chemotherapy following progression on immunotherapy in metastatic melanoma: an institutional case-series. Front. Oncol. 12, 855782 (2022).
    • 25. Saint-Jean M, Fronteau C, Peuvrel L et al. Chemotherapy efficacy after first-line immunotherapy in 18 advanced melanoma patients. Medicine (Baltimore) 99(29), e21329 (2020).
    • 26. Orouji A, Goerdt S, Utikal J. Systemic therapy of non-resectable metastatic melanoma. Cancers (Basel) 2(2), 955–969 (2010).
    • 27. Vera Aguilera J, Paludo J, Mcwilliams RR et al. Chemo-immunotherapy combination after PD-1 inhibitor failure improves clinical outcomes in metastatic melanoma patients. Melanoma Res. 30(4), 364–375 (2020).
    • 28. Hadash-Bengad R, Hajaj E, Klein S et al. Immunotherapy potentiates the effect of chemotherapy in metastatic melanoma – a retrospective study. Front. Oncol. 10, 70 (2020).
    • 29. Kas B, Talbot H, Ferrara R et al. Clarification of definitions of hyperprogressive disease during immunotherapy for non-small cell lung cancer. JAMA Oncol. 6(7), 1039–1046 (2020).
    • 30. Sevko A, Michels T, Vrohlings M et al. Antitumor effect of paclitaxel is mediated by inhibition of myeloid-derived suppressor cells and chronic inflammation in the spontaneous melanoma model. J. Immunol. 190(5), 2464–2471 (2013).
    • 31. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 21(8), 485–498 (2021).
    • 32. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9(3), 162–174 (2009).
    • 33. Kim C, Sano Y, Todorova K et al. The kinase p38 alpha serves cell type-specific inflammatory functions in skin injury and coordinates pro- and anti-inflammatory gene expression. Nat. Immunol. 9(9), 1019–1027 (2008).
    • 34. Gajewski TF. Failure at the effector phase: immune barriers at the level of the melanoma tumor microenvironment. Clin. Cancer Res. 13(18 Pt 1), 5256–5261 (2007).
    • 35. Zhao F, Falk C, Osen W, Kato M, Schadendorf D, Umansky V. Activation of p38 mitogen-activated protein kinase drives dendritic cells to become tolerogenic in ret transgenic mice spontaneously developing melanoma. Clin. Cancer Res. 15(13), 4382–4390 (2009).
    • 36. Kwak T, Wang F, Deng H et al. Distinct populations of immune-suppressive macrophages differentiate from monocytic myeloid-derived suppressor cells in cancer. Cell. Rep. 33(13), 108571 (2020).
    • 37. Wang S, Song R, Wang Z, Jing Z, Wang S, Ma J. S100A8/A9 in inflammation. Front. Immunol. 9, 1298 (2018).
    • 38. Inciarte-Mundo J, Frade-Sosa B, Sanmarti R. From bench to bedside: calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front. Immunol. 13, 1001025 (2022).
    • 39. Garg SK, Sun J, Kim Y et al. Dichotomous nitric oxide-dependent post-translational modifications of STAT1 are associated with ipilimumab benefits in melanoma. Cancers (Basel) 15(6), (2023).
    • 40. Markowitz J, Wang J, Vangundy Z et al. Nitric oxide mediated inhibition of antigen presentation from DCs to CD4(+) T cells in cancer and measurement of STAT1 nitration. Sci. Rep. 7(1), 15424 (2017).
    • 41. Yarlagadda K, Hassani J, Foote IP, Markowitz J. The role of nitric oxide in melanoma. Biochim. Biophys Acta Rev. Cancer 1868(2), 500–509 (2017).
    • 42. Zhou J, Nefedova Y, Lei A, Gabrilovich D. Neutrophils and PMN-MDSC: their biological role and interaction with stromal cells. Semin. Immunol. 35, 19–28 (2018).
    • 43. Reddy TP, Glynn SA, Billiar TR, Wink DA, Chang JC. Targeting nitric oxide: say NO to metastasis. Clin. Cancer Res. 29(10), 1855–1868 (2022).