301
|
Maio M, Lahn M, Di Giacomo AM, Covre A, Calabrò L, Ibrahim R, Fox B. A vision of immuno-oncology: the Siena think tank of the Italian network for tumor biotherapy (NIBIT) foundation. J Exp Clin Cancer Res 2021; 40:240. [PMID: 34301276 PMCID: PMC8298945 DOI: 10.1186/s13046-021-02023-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/18/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The yearly Think Tank Meeting of the Italian Network for Tumor Biotherapy (NIBIT) Foundation, brings together in Siena, Tuscany (Italy), experts in immuno-oncology to review the learnings from current immunotherapy treatments, and to propose new pre-clinical and clinical investigations in selected research areas. MAIN: While immunotherapies in non-small cell lung cancer and melanoma led to practice changing therapies, the same therapies had only modest benefit for patients with other malignancies, such as mesothelioma and glioblastoma. One way to improve on current immunotherapies is to alter the sequence of each combination agent. Matching the immunotherapy to the host's immune response may thus improve the activity of the current treatments. A second approach is to combine current immunotherapies with novel agents targeting complementary mechanisms. Identifying the appropriate novel agents may require different approaches than the traditional laboratory-based discovery work. For example, artificial intelligence-based research may help focusing the search for innovative and most promising combination partners. CONCLUSION Novel immunotherapies are needed in cancer patients with resistance to or relapse after current immunotherapeutic drugs. Such new treatments may include targeted agents or monoclonal antibodies to overcome the immune-suppressive tumor microenvironment. The mode of combining the novel treatments, including vaccines, needs to be matched to the patient's immune status for achieving the maximum benefit. In this scenario, specific attention should be also paid nowadays to the immune intersection between COVID-19 and cancer.
Collapse
Affiliation(s)
- Michele Maio
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci, 16, Siena, Italy.
- Italian Network for Tumor Bio-Immunotherapy Foundation Onlus, Siena, Italy.
| | - Michael Lahn
- iOnctura SA, Avenue Secheron 15, Geneva, Switzerland
| | - Anna Maria Di Giacomo
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci, 16, Siena, Italy
- Italian Network for Tumor Bio-Immunotherapy Foundation Onlus, Siena, Italy
| | - Alessia Covre
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci, 16, Siena, Italy
| | - Luana Calabrò
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci, 16, Siena, Italy
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, San Francisco, 94012, USA
| | - Bernard Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, 4805 NE Glisan St. Suite 2N35, Portland, OR, 97213, USA
| |
Collapse
|
302
|
Indini A, Roila F, Grossi F, Massi D, Mandalà M. Impact of Circulating and Tissue Biomarkers in Adjuvant and Neoadjuvant Therapy for High-Risk Melanoma: Ready for Prime Time? Am J Clin Dermatol 2021; 22:511-522. [PMID: 34036489 PMCID: PMC8200339 DOI: 10.1007/s40257-021-00608-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
The prognosis of patients with metastatic melanoma has substantially improved over the last years with the advent of novel treatment strategies, mainly immune checkpoint inhibitors and BRAF and MEK inhibitors. Given the survival benefit provided in the metastatic setting and the evidence from prospective clinical trials in the early stages, these drugs have been introduced as adjuvant therapies for high-risk resected stage III disease. Several studies have also investigated immune checkpoint inhibitors, as well as BRAF and MEK inhibitors, for neoadjuvant treatment of high-risk stage III melanoma, with preliminary evidence suggesting this could be a very promising approach in this setting. However, even with new strategies, the risk of disease recurrence varies widely among stage III patients, and no available biomarkers for predicting disease recurrence have been established to date. Improved risk stratification is particularly relevant in this setting to avoid unnecessary treatment for patients who have minimum risk of disease recurrence and to reduce toxicities and costs. Research for predictive and prognostic biomarkers in this setting is ongoing to potentially shed light on the complex interplay between the tumor and the host immune system, and to further personalize treatment. This review provides an insight into available data on circulating and tissue biomarkers, including the tumor microenvironment and associated gene signatures, and their predictive and prognostic role during neoadjuvant and adjuvant treatment for cutaneous high-risk melanoma patients.
Collapse
Affiliation(s)
- Alice Indini
- Medical Oncology Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fausto Roila
- Unit of Medical Oncology, Department of Surgery and Medicine, University of Perugia, Perugia, Italy
| | - Francesco Grossi
- Unit of Medical Oncology, Ospedale di Circolo e Fondazione Macchi, Università dell'Insubria, Varese, Italy
| | - Daniela Massi
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, Department of Surgery and Medicine, University of Perugia, Perugia, Italy.
| |
Collapse
|
303
|
Dimitriou F, Long G, Menzies A. Novel adjuvant options for cutaneous melanoma. Ann Oncol 2021; 32:854-865. [DOI: 10.1016/j.annonc.2021.03.198] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/13/2021] [Accepted: 03/09/2021] [Indexed: 01/10/2023] Open
|
304
|
Russell BL, Sooklal SA, Malindisa ST, Daka LJ, Ntwasa M. The Tumor Microenvironment Factors That Promote Resistance to Immune Checkpoint Blockade Therapy. Front Oncol 2021; 11:641428. [PMID: 34268109 PMCID: PMC8276693 DOI: 10.3389/fonc.2021.641428] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Through genetic and epigenetic alterations, cancer cells present the immune system with a diversity of antigens or neoantigens, which the organism must distinguish from self. The immune system responds to neoantigens by activating naïve T cells, which mount an anticancer cytotoxic response. T cell activation begins when the T cell receptor (TCR) interacts with the antigen, which is displayed by the major histocompatibility complex (MHC) on antigen-presenting cells (APCs). Subsequently, accessory stimulatory or inhibitory molecules transduce a secondary signal in concert with the TCR/antigen mediated stimulus. These molecules serve to modulate the activation signal's strength at the immune synapse. Therefore, the activation signal's optimum amplitude is maintained by a balance between the costimulatory and inhibitory signals. This system comprises the so-called immune checkpoints such as the programmed cell death (PD-1) and Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and is crucial for the maintenance of self-tolerance. Cancers often evade the intrinsic anti-tumor activity present in normal physiology primarily by the downregulation of T cell activation. The blockade of the immune checkpoint inhibitors using specific monoclonal antibodies has emerged as a potentially powerful anticancer therapy strategy. Several drugs have been approved mainly for solid tumors. However, it has emerged that there are innate and acquired mechanisms by which resistance is developed against these therapies. Some of these are tumor-intrinsic mechanisms, while others are tumor-extrinsic whereby the microenvironment may have innate or acquired resistance to checkpoint inhibitors. This review article will examine mechanisms by which resistance is mounted against immune checkpoint inhibitors focussing on anti-CTL4-A and anti-PD-1/PD-Ll since drugs targeting these checkpoints are the most developed.
Collapse
Affiliation(s)
- Bonnie L. Russell
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
- Innovation Hub, Buboo (Pty) Ltd, Pretoria, South Africa
| | - Selisha A. Sooklal
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
| | - Sibusiso T. Malindisa
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
| | | | - Monde Ntwasa
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
| |
Collapse
|
305
|
Abstract
PURPOSE OF REVIEW Sarcoidosis is a complex granulomatous disease of unknown cause. Several drug categories are able to induce a systemic granulomatous indistinguishable from sarcoidosis, known as drug-induced sarcoidosis-like reaction (DISR). This granulomatous inflammation can resolve if the medication is discontinued. In this review, we discuss recent literature on medication associated with DISR, possible pathophysiology, clinical features, and treatment. RECENT FINDINGS Recently, increasing reports on DISR have expanded the list of drugs associated with the systemic granulomatous eruption. Most reported drugs can be categorized as combination antiretroviral therapy, tumor necrosis factor-α antagonist, interferons, and immune checkpoint inhibitors, but reports on other drugs are also published. The proposed mechanism is enhancement of the aberrant immune response which results in systemic granuloma formation. It is currently not possible to know whether DISR represents a separate entity or is a triggered but 'true' sarcoidosis.As DISRs may cause minimal symptoms, treatment is not always necessary and the benefits of continuing the offending drug should be weighed against clinical symptoms and organ dysfunction. Treatment may involve immunosuppressive medication that is used for sarcoidosis treatment. SUMMARY In this article, we review recent insights in DISR: associated drug categories, clinical presentation, diagnosis, and treatment. Additionally, we discuss possible mechanisms of DISR which can add to our knowledge of sarcoidosis pathophysiology.
Collapse
|
306
|
Li Y, Zhang Y, Cao G, Zheng X, Sun C, Wei H, Tian Z, Xiao W, Sun R, Sun H. Blockade of checkpoint receptor PVRIG unleashes anti-tumor immunity of NK cells in murine and human solid tumors. J Hematol Oncol 2021; 14:100. [PMID: 34174928 PMCID: PMC8236157 DOI: 10.1186/s13045-021-01112-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/13/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Although checkpoint-based immunotherapy has shown exciting results in the treatment of tumors, around 70% of patients have experienced unresponsiveness. PVRIG is a recently identified immune checkpoint receptor and blockade of which could reverse T cell exhaustion to treat murine tumor; however, its therapeutic potential via NK cells in mice and human remains seldom reported. METHODS In this study, we used patient paraffin-embedded colon adenocarcinoma sections, various murine tumor models (MC38 colon cancer, MCA205 fibrosarcoma and LLC lung cancer), and human NK cell- or PBMC-reconstituted xenograft models (SW620 colon cancer) to investigate the effect of PVRIG on tumor progression. RESULTS We found that PVRIG was highly expressed on tumor-infiltrating NK cells with exhausted phenotype. Furthermore, either PVRIG deficiency, early blockade or late blockade of PVRIG slowed tumor growth and prolonged survival of tumor-bearing mice by inhibiting exhaustion of NK cells as well as CD8+ T cells. Combined blockade of PVRIG and PD-L1 showed better effect in controlling tumor growth than using either one alone. Depletion of NK or/and CD8+ T cells in vivo showed that both cell types contributed to the anti-tumor efficacy of PVRIG blockade. By using Rag1-/- mice, we demonstrated that PVRIG blockade could provide therapeutic effect in the absence of adaptive immunity. Further, blockade of human PVRIG with monoclonal antibody enhanced human NK cell function and inhibited human tumor growth in NK cell- or PBMC-reconstituted xenograft mice. CONCLUSIONS Our results reveal the importance of NK cells and provide novel knowledge for clinical application of PVRIG-targeted drugs in future.
Collapse
Affiliation(s)
- Yangyang Li
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yu Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Guoshuai Cao
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiaodong Zheng
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Cheng Sun
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing, China
| | - Weihua Xiao
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Hefei TG ImmunoPharma Corporation Limited, Hefei, China
| | - Rui Sun
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, China.
| | - Haoyu Sun
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 443 Huangshan Road, Hefei, 230027, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
307
|
A Pilot Study of Short-course Nivolumab and Low-dose Ipilimumab for Adjuvant Treatment of Melanoma: Brown University Oncology Research Group Trial, BrUOG 324. Am J Clin Oncol 2021; 44:254-257. [PMID: 33899806 DOI: 10.1097/coc.0000000000000820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Combined cytotoxic T-lymphocyte-associated antigen 4 and programmed death 1 inhibitor blockade is a promising strategy in advanced melanoma and other solid tumors. This pilot study assessed the safety and toxicity of nivolumab plus low-dose ipilimumab in patients with high-risk completely resected melanoma. PATIENTS AND METHODS Patients received ipilimumab, 1 mg/kg every 6 weeks, and nivolumab, 3 mg/kg every 2 weeks, for a total of 24 weeks (4 cycles). The primary objective was to assess the toxicity of the combined regimen. RESULTS Twenty-one patients with resected melanoma were enrolled. One patient was stage IIC, 16 patients were stage III and 4 patients had resected stage 4 disease. Ten of 21 (48%) had grade 3 treatment-related toxicities but there was no grade 4 or grade 5 toxicities. The rate of grade 3 nonhematologic toxicities exceeded the toxicity limits defined by the study. Fifteen of 21 patients (71%) completed all 4 cycles of therapy. The median follow-up is 41 months. The 2-year recurrence-free survival is 85.7% and the 2-year overall survival is 90.5%. CONCLUSION A 6-month course of nivolumab and low-dose ipilimumab may be a promising adjuvant treatment for patients with resected melanoma. Further studies of this regimen are indicated.
Collapse
|
308
|
Turchan WT, Pitroda SP, Weichselbaum RR. Radiotherapy and Immunotherapy Combinations in the Treatment of Patients with Metastatic Disease: Current Status and Future Focus. Clin Cancer Res 2021; 27:5188-5194. [PMID: 34140404 DOI: 10.1158/1078-0432.ccr-21-0145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/09/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022]
Abstract
Radiotherapy and immunotherapy benefit subsets of patients with metastatic cancer. Here, we review selected laboratory and clinical studies investigating the utility of combining radiotherapy and immunotherapy in metastatic patients. We examine potential approaches to increase the therapeutic ratio of radioimmunotherapy in the treatment of metastatic cancers moving forward.
Collapse
Affiliation(s)
- William Tyler Turchan
- University of Chicago, Department of Radiation and Cellular Oncology, Chicago, Illinois
| | - Sean P Pitroda
- University of Chicago, Department of Radiation and Cellular Oncology, Chicago, Illinois
| | - Ralph R Weichselbaum
- University of Chicago, Department of Radiation and Cellular Oncology, Chicago, Illinois.
| |
Collapse
|
309
|
Mojtahed SA, Boyer NR, Rao SA, Gajewski TF, Tseng J, Turaga KK. Cost-Effectiveness Analysis of Adjuvant Therapy for BRAF-Mutant Resected Stage III Melanoma in Medicare Patients. Ann Surg Oncol 2021; 28:9039-9047. [PMID: 34129153 DOI: 10.1245/s10434-021-10288-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/23/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Adjuvant therapy for stage III melanoma improves several measures of patient survival. However, decisions regarding inclusion of adjuvant therapies in the formularies of public payers necessarily consider the cost-effectiveness of those treatments. The objective of this study is to evaluate the cost-effectiveness of four recently approved adjuvant therapies for BRAF-mutant stage III melanoma in the Medicare patient population. METHODS In this cost-effectiveness analysis, a Markov microsimulation model was used to simulate the healthcare trajectory of patients randomized to receive either first-line targeted therapy (dabrafenib-trametinib) or immunotherapy (ipilimumab, nivolumab, or pembrolizumab). The base case was a 65-year-old Medicare patient with BRAF V600E-mutant resected stage III melanoma. Possible health states included recurrence-free survival, adverse events, local recurrence, distant metastases, and death. Transition probabilities were determined from published clinical trials. Costs were estimated from reimbursement rates reported by CMS and the Red Book drug price database. Primary outcomes were costs (US$), life years, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs). Model robustness was evaluated using one-way and probabilistic sensitivity analyses. RESULTS Dabrafenib-trametinib provided 1.83 QALYs over no treatment and 0.23 QALYs over the most effective immunotherapy, pembrolizumab. Dabrafenib-trametinib was associated with an ICER of $95,758/QALY over no treatment and $285,863/QALY over pembrolizumab. Pembrolizumab yielded an ICER of $68,396/QALY over no treatment and dominated other immunotherapies. CONCLUSIONS Pembrolizumab is cost-effective at a conventional willingness-to-pay (WTP) threshold, but dabrafenib-trametinib is not. Though dabrafenib-trametinib offers incremental QALYs, optimization of drug pricing is necessary to ensure dabrafenib-trametinib is accessible at an acceptable WTP threshold.
Collapse
Affiliation(s)
- Saam A Mojtahed
- Pritzker School of Medicine, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| | - Nicole R Boyer
- Center for Health and the Social Sciences, University of Chicago, Chicago, IL, USA
| | - Saieesh A Rao
- Pritzker School of Medicine, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| | - Thomas F Gajewski
- Department of Pathology, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| | - Jennifer Tseng
- Department of Surgery, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Kiran K Turaga
- Department of Surgery, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
310
|
Bergamini C, Ferris RL, Xie J, Mariani G, Ali M, Holmes WC, Harrington K, Psyrri A, Cavalieri S, Licitra L. Bleeding complications in patients with squamous cell carcinoma of the head and neck. Head Neck 2021; 43:2844-2858. [PMID: 34117666 PMCID: PMC8453784 DOI: 10.1002/hed.26772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/17/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Hemorrhage in recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) may be attributed to chemotherapy and local tumor irradiation. Evidence of the relationship between hemorrhage in R/M HNSCC and targeted therapies, including epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) inhibitors, or immune checkpoint inhibitors, is limited. We aimed to identify epidemiological and clinical data related to the occurrence of hemorrhage in R/M HNSCC and to explore its relationship with various therapies. We describe information obtained from literature searches as well as data extracted from a commercial database and a database from the author's institution (Istituto Nazionale dei Tumori of Milan). Evidence suggests that most bleeding events in R/M HNSCC are minor. Clinical trial safety data do not identify a causal association between hemorrhage and anti‐EGFR agents or immune checkpoint inhibitors. In contrast, anti‐VEGF agents are associated with increased, and often severe/fatal, hemorrhagic complications.
Collapse
Affiliation(s)
- Cristiana Bergamini
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Robert L Ferris
- Department of Otolaryngology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jing Xie
- Department of Epidemiology, AstraZeneca, Gaithersburg, Maryland, USA
| | | | - Muzammil Ali
- Global Medicine Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - William C Holmes
- Global Medicine Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Kevin Harrington
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, National Institute of Health Research Biomedical Research Centre, London, UK
| | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Italy
| |
Collapse
|
311
|
Affiliation(s)
- Brendan D Curti
- From the Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR (B.D.C.); and Cedars-Sinai Medical Center and the Angeles Clinic and Research Institute, Los Angeles (M.B.F.)
| | - Mark B Faries
- From the Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR (B.D.C.); and Cedars-Sinai Medical Center and the Angeles Clinic and Research Institute, Los Angeles (M.B.F.)
| |
Collapse
|
312
|
Park R, Park JC. Current landscape of immunotherapy trials in locally advanced and high-risk head and neck cancer. Immunotherapy 2021; 13:931-940. [PMID: 34100301 DOI: 10.2217/imt-2021-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The current standard of therapy for locally advanced (LA) head and neck squamous cell carcinoma (HNSCC) is limited by toxicity and suboptimal control. The role of immunotherapy (IO) is being evaluated in the LA setting. This review aims to summarize the recent advances and the direction of clinical trials in IO in LA or high-risk HNSCC. Despite negative results in some studies, several early phase trials suggest the feasibility and efficacy of IO-based strategies in LA or high-risk HNSCC. Further refining of patient selection and biomarker development is warranted for successful incorporation of IO in this patient population.
Collapse
Affiliation(s)
- Robin Park
- Department of Medicine, MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA 01702, USA
| | - Jong Chul Park
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, Boston, MA 02114, USA.,Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
313
|
Maio M, Blank C, Necchi A, Di Giacomo AM, Ibrahim R, Lahn M, Fox BA, Bell RB, Tortora G, Eggermont AMM. Neoadjuvant immunotherapy is reshaping cancer management across multiple tumour types: The future is now! Eur J Cancer 2021; 152:155-164. [PMID: 34107449 DOI: 10.1016/j.ejca.2021.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022]
Abstract
The Italian Network for Tumor Biotherapy (Network Italiano per la Bioterapia dei Tumori [NIBIT]) Foundation hosted its annual 2020 Think Tank meeting virtually, at which representatives from academic, clinical, industry, philanthropic, and regulatory organisations discussed the role of neoadjuvant immunotherapy for the treatment of cancer. Although the number of neoadjuvant immunotherapeutic trials is increasing across all malignancies, the Think Tank focused its discussion on the status of neoadjuvant trials in cutaneous melanoma (CM), muscle-invasive urothelial bladder cancer (MIBC), head and neck squamous cell carcinoma (HNSCC), and pancreatic adenocarcinoma (PDAC). Neoadjuvant developments in CM are nothing short of trailblazing. Pathologic Complete Response (pCR), pathologic near Complete Response, and partial Pathologic Responses reduce 90-100% of recurrences. This is in sharp contrast to targeted therapies in neoadjuvant CM trials, where only a pCR seems to reduce recurrence. The pCR rate after neoadjuvant immunotherapy varies among the different malignancies of CM, MIBC, HNSCC, and PDAC and may be associated with different reductions of recurrence rates. In CM, emerging evidence suggests that neoadjuvant immunotherapy with anti-CTLA-4 plus anti-PD1 is a game changer in patients with palpable nodal Stage III or resectable Stage IV disease by curing more patients, reducing recurrences and the need for surgical interventions, such as lymph node dissections and metastasectomies. The Think Tank panel discussed future approaches on how to optimise results across different tumour types. Future approaches should include reducing monocyte-mediated (tumour-associated macrophages) and fibroblast-mediated (cancer-associated fibroblasts) barriers in the tumour microenvironment to facilitate the recruitment of immune cells to the tumour site, to reduce immune-suppressive mediators, and to increase antigen presentation at the site of the tumour.
Collapse
Affiliation(s)
- Michele Maio
- Center for Immuno-Oncology, Department of Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci 16, Siena, Italy; Italian Network for Tumor Bio-Immunotherapy Foundation, Siena, Italy.
| | - Christian Blank
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| | - Andrea Necchi
- Genitourinary Medical Oncology, Vita-Salute San Raffaele University, Via Olgettina 60, 20132 Milan, Italy.
| | - Anna Maria Di Giacomo
- Center for Immuno-Oncology, Department of Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci 16, Siena, Italy; Italian Network for Tumor Bio-Immunotherapy Foundation, Siena, Italy.
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| | - Michael Lahn
- IOnctura SA, Avenue Secheron 15, Geneva, Switzerland.
| | - Bernard A Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Providence Portland Medical Center, 4805 NE Glisan, Portland, OR 97213, USA.
| | - R Bryan Bell
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Providence Portland Medical Center, 4805 NE Glisan, Portland, OR 97213, USA.
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Gemelli IRCCS e Università Cattolica Del Sacro Cuore, Roma, Largo Agostino Gemelli 8, 00168 Roma, Italy.
| | - Alexander M M Eggermont
- Princess Máxima Center, University Medical Center Utrecht, Heidelberglaan 25, 3584 Utrecht, the Netherlands.
| |
Collapse
|
314
|
De Santis F, Fucà G, Schadendorf D, Mantovani A, Magnani L, Lisanti M, Pettitt S, Bellone M, Del Sal G, Minucci S, Eggermont A, Bruzzi P, Bicciato S, Conte P, Noberini R, Hiscott J, De Braud F, Del Vecchio M, Di Nicola M. Anticancer innovative therapy congress: Highlights from the 10th anniversary edition. Cytokine Growth Factor Rev 2021; 59:1-8. [PMID: 33610464 DOI: 10.1016/j.cytogfr.2021.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
During the Tenth Edition of the Annual Congress on "Anticancer Innovative Therapy" [Milan, 23/24 January 2020], experts in the fields of immuno-oncology, epigenetics, tumor cell signaling, and cancer metabolism shared their latest knowledge on the roles of i] epigenetics, and in particular, chromatin modifiers, ii] cancer metabolism, iii] cancer stem cells [CSCs], iv] tumor cell signaling, and iv] the immune system. The novel therapeutic approaches presented included epigenetic drugs, cell cycle inhibitors combined with ICB, antibiotics and other off-label drugs, small-molecules active against CSCs, liposome-delivered miRNAs, tumor-specific CAR-T cells, and T-cell-based immunotherapy. Moreover, important evidence on possible mechanisms of resistance to these innovative therapies were also discussed, in particular with respect to resistance to ICB. Overall, this conference provided scientists and clinicians with a broad overview of future challenges and hopes to improve cancer treatment reasonably in the medium-short term.
Collapse
Affiliation(s)
- Francesca De Santis
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | | | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Michael Lisanti
- Translational Medicine, School of Science, Engineering and Environment [SEE], Biomedical Research Centre [BRC], University of Salford, Greater Manchester, United Kingdom
| | - Stephen Pettitt
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom
| | - Matteo Bellone
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alexander Eggermont
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, the Netherlands
| | - Paolo Bruzzi
- Unit of Clinical Epidemiology, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pierfranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Noberini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - John Hiscott
- Laboratorio Pasteur, Istituto Pasteur-Fondazione Cenci-Bolognetti, 00161, Rome, Italy
| | - Filippo De Braud
- Department of Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Del Vecchio
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Di Nicola
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
315
|
Fang T, Xiao J, Zhang Y, Hu H, Zhu Y, Cheng Y. Combined with interventional therapy, immunotherapy can create a new outlook for tumor treatment. Quant Imaging Med Surg 2021; 11:2837-2860. [PMID: 34079746 PMCID: PMC8107298 DOI: 10.21037/qims-20-173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Recent progress in immunotherapy provides hope of a complete cure to cancer patients. However, recent studies have reported that only a limited number of cancer patients with a specific immune status, known as "cold tumor", can benefit from a single immune agent. Although the combination of immune agents with different mechanisms can partially increase the low response rate and improve efficacy, it can also result in more side effects. Therefore, discovering therapies that can improve tumors' response rate to immunotherapy without increasing toxicity for patients is urgently needed. Tumor interventional therapy is promising. It mainly includes transcatheter arterial chemoembolization, ablation, radioactive particle internal irradiation, and photodynamic interventional therapy based on a luminal stent. Interventional therapy can directly kill tumor cells by targeted drug delivery in situ, thus reducing drug dosage and systemic toxicity like cytokine release syndrome. More importantly, interventional therapy can regulate the immune system through numerous mechanisms, making it a suitable choice for immunotherapy to combine with. In this review, we provide a brief description of immunotherapies (and their side effects) on tumors of different immune types and preliminarily elaborate on interventional therapy mechanisms to improve immune efficacy. We also discuss the progress and challenges of the combination of interventional therapy and immunotherapy.
Collapse
Affiliation(s)
- Tonglei Fang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Junyuan Xiao
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yiran Zhang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Haiyan Hu
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yueqi Zhu
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yingsheng Cheng
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
316
|
Malvi P, Janostiak R, Nagarajan A, Zhang X, Wajapeyee N. N-acylsphingosine amidohydrolase 1 promotes melanoma growth and metastasis by suppressing peroxisome biogenesis-induced ROS production. Mol Metab 2021; 48:101217. [PMID: 33766731 PMCID: PMC8081993 DOI: 10.1016/j.molmet.2021.101217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/02/2021] [Accepted: 03/17/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Metabolic deregulation is a key hallmark of cancer cells and has been shown to drive cancer growth and metastasis. However, not all metabolic drivers of melanoma are known. Based on our finding that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in melanoma, the objective of these studies was to establish its role in melanoma tumor growth and metastasis, understand its mechanism of action, and evaluate ASAH1 targeting for melanoma therapy. METHODS We used publicly available melanoma datasets and patient-derived samples of melanoma and normal skin tissue and analyzed them for ASAH1 mRNA expression and ASAH1 protein expression using immunohistochemistry. ASAH1 was knocked down using short-hairpin RNAs in multiple melanoma cell lines that were tested in a series of cell culture-based assays and mouse-based melanoma xenograft assays to monitor the effect of ASAH1 knockdown on melanoma tumor growth and metastasis. An unbiased metabolomics analysis was performed to identify the mechanism of ASAH1 action. Based on the metabolomics findings, the role of peroxisome-mediated reactive oxygen species (ROS) production was explored in regard to mediating the effect of ASAH1. The ASAH1 inhibitor was used alone or in combination with a BRAFV600E inhibitor to evaluate the therapeutic value of ASAH1 targeting for melanoma therapy. RESULTS We determined that ASAH1 was overexpressed in a large percentage of melanoma cells and regulated by transcription factor E2F1 in a mitogen-activated protein (MAP) kinase pathway-dependent manner. ASAH1 expression was necessary to maintain melanoma tumor growth and metastatic attributes in cell cultures and mouse models of melanoma tumor growth and metastasis. To identify the mechanism by which ASAH1 facilitates melanoma tumor growth and metastasis, we performed a large-scale and unbiased metabolomics analysis of melanoma cells expressing ASAH1 short-hairpin RNAs (shRNAs). We found that ASAH1 inhibition increased peroxisome biogenesis through ceramide-mediated PPARγ activation. ASAH1 loss increased ceramide and peroxisome-derived ROS, which in turn inhibited melanoma growth. Pharmacological inhibition of ASAH1 also attenuated melanoma growth and enhanced the effectiveness of BRAF kinase inhibitor in the cell cultures and mice. CONCLUSIONS Collectively, these results demonstrate that ASAH1 is a druggable driver of melanoma tumor growth and metastasis that functions by suppressing peroxisome biogenesis, thereby inhibiting peroxisome-derived ROS production. These studies also highlight the therapeutic utility of ASAH1 inhibitors for melanoma therapy.
Collapse
Affiliation(s)
- Parmanand Malvi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama, 35233, USA
| | - Radoslav Janostiak
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Arvindhan Nagarajan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama, 35233, USA.
| |
Collapse
|
317
|
Vuoristo M, Muhonen T, Koljonen V, Juteau S, Hernberg M, Ilmonen S, Jahkola T. Long-term prognostic value of sentinel lymph node tumor burden in survival of melanoma patients. Acta Oncol 2021; 60:803-807. [PMID: 33656957 DOI: 10.1080/0284186x.2021.1892820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Mikko Vuoristo
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Timo Muhonen
- Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Virve Koljonen
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Susanna Juteau
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Micaela Hernberg
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Oncology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Suvi Ilmonen
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tiina Jahkola
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
318
|
Nebhan CA, Johnson DB. Pembrolizumab in the adjuvant treatment of melanoma: efficacy and safety. Expert Rev Anticancer Ther 2021; 21:583-590. [PMID: 33504219 PMCID: PMC8238788 DOI: 10.1080/14737140.2021.1882856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Introduction: Regional or distant metastases from melanoma may be surgically resected but remain at high-risk of recurrence. Over the last few years, several treatments have been approved to mitigate this risk. These include anti-PD-1 agents, specifically pembrolizumab and nivolumab.Areas covered: Herein, we will discuss the landscape of pembrolizumab safety and efficacy used in the adjuvant setting for high-risk, resected melanoma. We place this in context with other available adjuvant therapies, and discuss subgroup analyses.Expert opinion: Anti-PD-1 therapy with either pembrolizumab or nivolumab has become a standard of care for patients with resected stage III or IV melanoma. In our practice, we generally offer these agents (which have comparable safety and efficacy profiles) to patients with resected stage IIIb-IV melanoma regardless of BRAF mutation status.
Collapse
Affiliation(s)
- Caroline A. Nebhan
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center
| |
Collapse
|
319
|
Wurcel V, Scherrer E, Aguiar-Ibanez R, Altuna JI, Carabajal F, Jain S, Baluni G. Cost-Effectiveness of Pembrolizumab for the Adjuvant Treatment of Melanoma Patients with Lymph Node Involvement Who Have Undergone Complete Resection in Argentina. Oncol Ther 2021; 9:167-185. [PMID: 33624271 PMCID: PMC8140053 DOI: 10.1007/s40487-021-00142-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/23/2021] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION The KEYNOTE-054 trial demonstrated that adjuvant pembrolizumab improves recurrence-free survival in completely resected stage III melanoma versus watchful waiting (hazard ratio [HR] = 0.57; 98.4% confidence interval [CI], 0.43-0.74). We evaluated the cost-effectiveness of pembrolizumab in Argentina, where watchful waiting is still widely used among these patients despite the high risk of recurrence with surgery alone. METHODS A four-health state model was used (recurrence-free, locoregional recurrence [LR], distant metastases [DM], death). Lifetime medical costs to payers (72.08 Argentine pesos [AR$] = 1.00 U.S. dollar [USD]) and outcomes (3% annual discount) were assessed, together with incremental cost-effectiveness ratios (ICERs). First and LR→DM recurrences were modeled using KEYNOTE-054 and real-world data, respectively. No benefits of adjuvant treatment were assumed post-progression. Pre-DM and post-DM mortality was based on KEYNOTE-054 and on a network meta-analysis of advanced treatments expected in each arm, respectively. Utilities were derived from KEYNOTE-054 Euro-QoL data using an Argentinian algorithm, and from the literature. Public ex-factory drug prices were used. RESULTS Patients in the pembrolizumab and the watchful waiting arms accrued 8.78 and 5.83 quality-adjusted life-years (QALYs), 9.91 and 6.98 life-years, and costs of AR$12,698,595 (176,174 USD) and AR$11,967,717 (166,034 USD), respectively. The proportion of life-years accrued that were recurrence-free was 80.8% and 56.9% in the pembrolizumab and the watchful waiting arms, respectively. Pembrolizumab patients gained 2.94 life-years and 2.96 QALYs versus watchful waiting; the ICER per QALY was AR$247,094 (3428 USD). Recurrence rates and advanced melanoma treatments were the key drivers of the ICER. At a threshold of AR$1,445,325 (29,935 USD) per QALY, pembrolizumab had an 83.5% probability of being cost-effective versus watchful waiting. CONCLUSIONS Adjuvant pembrolizumab after complete resection of melanoma with node involvement is highly cost-effective relative to watchful waiting in Argentina, across disease stage subgroups and BRAF mutational status. This strongly supports its coverage and reimbursement across the entire health system.
Collapse
Affiliation(s)
| | | | | | | | | | - Shrishti Jain
- Complete HEOR Solutions, CHEORS, North Wales, PA, USA
| | - Gargi Baluni
- Complete HEOR Solutions, CHEORS, North Wales, PA, USA
| |
Collapse
|
320
|
Atieh J, Sack J, Thomas R, Rahma OE, Camilleri M, Grover S. Gastroparesis Following Immune Checkpoint Inhibitor Therapy: A Case Series. Dig Dis Sci 2021; 66:1974-1980. [PMID: 32594464 PMCID: PMC7867661 DOI: 10.1007/s10620-020-06440-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/21/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have improved outcomes in patients with various malignancies; however, they can cause immune-related hepatitis and enterocolitis. Patients on ICI may also develop upper gastrointestinal symptoms and undergo measurement of gastric emptying. AIMS Our aim was to review records of patients with gastroparesis following ICI therapy at two medical centers. METHODS We performed a retrospective review of all patients at Mayo Clinic and Brigham and Women's/Dana-Farber Cancer Center (BWH/DFCC) who underwent gastric scintigraphy for the assessment of symptoms of gastroparesis following ICI treatment up to January 2020. Clinical presentation, medical history, laboratory evaluation, imaging, treatment, and outcomes were retrieved from the records. Gastroparesis was diagnosed as delayed gastric emptying (GE) measured by gastric scintigraphy. RESULTS At Mayo Clinic, 2 patients (median age 59 years, 1 male [M], 1 female [F]) had delayed GE, while 4 patients (median age 53 years, 3M, 1F) had normal GE following ICI use. Of those with delayed GE (diagnosed after 38 and 2 months of ICI initiation), 1 patient was treated for non-Hodgkin's lymphoma and melanoma with ipilimumab; a second patient with breast cancer was treated with pembrolizumab. At BWH/DFCC, 2 patients (median age 56 years, 1M, 1F) had normal GE after ICI treatment, while a 62-year-old female with non-small cell lung cancer developed gastroparesis 3 months following initiation of nivolumab. CONCLUSION This report documents gastroparesis as a potential adverse effect of ICI. Further studies should explore the potential for ICI therapy to damage anti-inflammatory macrophages that preserve the enteric neurons.
Collapse
Affiliation(s)
- Jessica Atieh
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER) and Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St. S.W., Rochester, MN, USA
| | - Jordan Sack
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Richard Thomas
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Osama E Rahma
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brookline, MA, USA
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER) and Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St. S.W., Rochester, MN, USA.
| | - Shilpa Grover
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
321
|
Egger ME. The Role of Clinical Prediction Tools to Risk Stratify Patients with Melanoma After a Positive Sentinel Lymph Node Biopsy. Ann Surg Oncol 2021; 28:4082-4083. [PMID: 34047858 DOI: 10.1245/s10434-018-07099-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Michael E Egger
- Hiram C. Polk Jr., M.D. Department of Surgery, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
322
|
Ascierto PA, Del Vecchio M, Mackiewicz A, Robert C, Chiarion-Sileni V, Arance A, Lebbé C, Svane IM, McNeil C, Rutkowski P, Loquai C, Mortier L, Hamid O, Bastholt L, Dreno B, Schadendorf D, Garbe C, Nyakas M, Grob JJ, Thomas L, Liszkay G, Smylie M, Hoeller C, Ferraresi V, Grange F, Gutzmer R, Pikiel J, Hosein F, Simsek B, Maio M. Overall survival at 5 years of follow-up in a phase III trial comparing ipilimumab 10 mg/kg with 3 mg/kg in patients with advanced melanoma. J Immunother Cancer 2021; 8:jitc-2019-000391. [PMID: 32503946 PMCID: PMC7279645 DOI: 10.1136/jitc-2019-000391] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We have previously reported significantly longer overall survival (OS) with ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with advanced melanoma, with higher incidences of adverse events (AEs) at 10 mg/kg. This follow-up analysis reports a 5-year update of OS and safety. METHODS This randomized, multicenter, double-blind, phase III trial included patients with untreated or previously treated unresectable stage III or IV melanoma. Patients were randomly assigned (1:1) to ipilimumab 10 mg/kg or 3 mg/kg every 3 weeks for 4 doses. The primary end point was OS. RESULTS At a minimum follow-up of 61 months, median OS was 15.7 months (95% CI 11.6 to 17.8) at 10 mg/kg and 11.5 months (95% CI 9.9 to 13.3) at 3 mg/kg (HR 0.84, 95% CI 0.71 to 0.99; p=0.04). In a subgroup analysis, median OS of patients with asymptomatic brain metastasis was 7.0 months (95% CI 4.0 to 12.8) in the 10 mg/kg group and 5.7 months (95% CI 4.2 to 7.0) in the 3 mg/kg group. In patients with wild-type or mutant BRAF tumors, median OS was 13.8 months (95% CI 10.2 to 17.0) and 33.2 months (95% CI 19.4 to 45.2) in the 10 mg/kg group, and 11.2 months (95% CI 9.2 to 13.8) and 19.7 months (95% CI 11.6 to 25.3) in the 3 mg/kg group, respectively. The incidence of grade 3/4 treatment-related AEs was 36% in the 10 mg/kg group vs 20% in the 3 mg/kg group, and deaths due to treatment-related AEs occurred in four (1%) and two patients (1%), respectively. CONCLUSIONS This 61-month follow-up of a phase III trial showed sustained long-term survival in patients with advanced melanoma who started metastatic treatment with ipilimumab monotherapy, and confirmed the significant benefit for those who received ipilimumab 10 mg/kg vs 3 mg/kg. These results suggest the emergence of a plateau in the OS curve, consistent with previous ipilimumab studies. TRIAL REGISTRATION NUMBER NCT01515189.
Collapse
Affiliation(s)
- Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Michele Del Vecchio
- Unit of Melanoma Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Andrzej Mackiewicz
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Center, Poznan Medical University, Poznan, Poland
| | - Caroline Robert
- Department of Medicine, Dermatology Service, Gustave Roussy, Villejuif and Paris-Sud-University, Le Kremlin-Bicêtre, France
| | | | - Ana Arance
- Hospital Clinic and Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Céleste Lebbé
- Université de Paris, INSERM, Dermatology and CIC, Saint Louis Hospital, Paris, France
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Herlev Hospital, Herlev, Denmark.,Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Catriona McNeil
- Chris O'Brien Lifehouse and Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Carmen Loquai
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Laurent Mortier
- Clinique de Dermatologie, Unité d'Onco-Dermatologie, INSERM U1189, Centre Hospitalier Régional Universitaire de Lille, Hôpital Claude Huriez, Lille, France
| | - Omid Hamid
- Melanoma Center, The Angeles Clinic and Research Institute, Los Angeles, California, USA
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Brigitte Dreno
- Department of Oncodermatology, University Hospital Centre Nantes, Nantes, Pays de la Loire, France
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany.,Department of Dermatology, German Cancer Consortium, Heidelberg, Germany
| | - Claus Garbe
- Department of Dermatology, Eberhard Karls Universitat Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Marta Nyakas
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jean-Jacques Grob
- Dermatology and Skin Cancers Department, Aix-Marseille University, APHM, Marseille, France
| | - Luc Thomas
- Department of Dermatology, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Gabriella Liszkay
- Department of Oncodermatology, National Institute of Oncology, Budapest, Hungary
| | - Michael Smylie
- Department of Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Christoph Hoeller
- Division of General Dermatology and Dermato-Oncology, Medical University of Vienna, Vienna, Austria
| | - Virginia Ferraresi
- Unit of Medical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Florent Grange
- Department of Dermatology, University Hospital Centre Reims, Reims, Champagne-Ardenne, France
| | - Ralf Gutzmer
- Operative Dermatology and Dermato-Oncology, Medizinische Hochschule Hannover, Hannover, Niedersachsen, Germany
| | - Joanna Pikiel
- Department of Oncology, Wojewodzkie Centrum Oncologii, Gdańsk, Poland
| | - Fareeda Hosein
- Oncology Clinical Development, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Burcin Simsek
- Oncology Clinical Development, Bristol-Myers Squibb Co, Princeton, New Jersey, USA.,Department of Biostatistics, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Michele Maio
- Center for Immuno-Oncology, University Hospital of Siena, Instituto Toscano Tumori, Siena, Italy
| |
Collapse
|
323
|
Patel A, Carr MJ, Sun J, Zager JS. In-transit metastatic cutaneous melanoma: current management and future directions. Clin Exp Metastasis 2021; 39:201-211. [PMID: 33999365 DOI: 10.1007/s10585-021-10100-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
Management of in-transit melanoma encompasses a variety of possible treatment pathways and modalities. Depending on the location of disease, number of lesions, burden of disease and patient preference and characteristics, some treatments may be more beneficial than others. After full body radiographic staging is performed to rule out metastatic disease, curative therapy may be performed through surgical excision, intraarterial regional perfusion and infusion therapies, intralesional injections, systemic therapies or various combinations of any of these. While wide excision is limited in indication to superficial lesions that are few in number, the other listed therapies may be effective in treating unresectable disease. Where intraarterial perfusion based therapies have been shown to successfully treat extremity disease, injectable therapies can be used in lesions of the head and neck. Although systemic therapies for in-transit melanoma have limited specific data to support their primary use for in-transit disease, there are patients who may not be eligible for any of the other options, and current clinical trials are exploring the use of concurrent and sequential use of regional and systemic therapies with early results suggesting a synergistic benefit for oncologic response and outcomes.
Collapse
Affiliation(s)
- Ayushi Patel
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Michael J Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA
| | - James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA.,Department of Surgery, University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Zager
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA. .,Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
324
|
Chick RC, Faries MB, Hale DF, Kemp Bohan PM, Hickerson AT, Vreeland TJ, Myers JW, Cindass JL, Brown TA, Hyngstrom J, Berger AC, Jakub JW, Sussman JJ, Shaheen M, Clifton GT, Park H, Sloan AJ, Wagner T, Peoples GE. Multi-institutional, prospective, randomized, double-blind, placebo-controlled phase IIb trial of the tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine to prevent recurrence in high-risk melanoma patients: A subgroup analysis. Cancer Med 2021; 10:4302-4311. [PMID: 33982452 PMCID: PMC8267143 DOI: 10.1002/cam4.3969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022] Open
Abstract
Background Checkpoint inhibitors (CPI) in combination with cell‐based vaccines may produce synergistic antitumor immunity. The primary analysis of the randomized and blinded phase IIb trial in resected stage III/IV melanoma demonstrated TLPLDC is safe and improved 24‐month disease‐free survival (DFS) in the per treatment (PT) analysis. Here, we examine efficacy within pre‐specified and exploratory subgroups. Methods Stage III/IV patients rendered disease‐free by surgery were randomized 2:1 to TLPLDC vaccine versus placebo. The pre‐specified PT analysis included only patients completing the primary vaccine/placebo series at 6 months. Kaplan–Meier analysis was used to compare 24‐month DFS among subgroups. Results There were no clinicopathologic differences between subgroups except stage IV patients were more likely to receive CPI. In stage IV patients, 24‐month DFS was 43% for vaccine versus 0% for placebo (p = 0.098) in the ITT analysis and 73% versus 0% (p = 0.002) in the PT analysis. There was no significant difference in 24‐month DFS when stratified by use of immunotherapy or CPI. For patients with resected recurrent disease, 24‐month DFS was 88.9% versus 33.3% (p = 0.013) in the PT analysis. All benefit from vaccination was in the PT analysis; no benefit was found in patients receiving up to three doses. Conclusion The TLPLDC vaccine improved DFS in patients completing the primary vaccine series, particularly in the resected stage IV patients. The efficacy of the TLPLDC vaccine will be confirmed in a phase III study evaluating adjuvant TLPLDC + CPI versus Placebo + CPI in resected stage IV melanoma patients.
Collapse
Affiliation(s)
| | | | - Diane F Hale
- Brooke Army Medical Center, Fort Sam Houston, TX, USA
| | | | | | | | - John W Myers
- Brooke Army Medical Center, Fort Sam Houston, TX, USA
| | | | - Tommy A Brown
- Brooke Army Medical Center, Fort Sam Houston, TX, USA
| | - John Hyngstrom
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Adam C Berger
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | | | | | - Guy T Clifton
- Brooke Army Medical Center, Fort Sam Houston, TX, USA
| | | | | | | | | |
Collapse
|
325
|
Munari E, Mariotti FR, Quatrini L, Bertoglio P, Tumino N, Vacca P, Eccher A, Ciompi F, Brunelli M, Martignoni G, Bogina G, Moretta L. PD-1/PD-L1 in Cancer: Pathophysiological, Diagnostic and Therapeutic Aspects. Int J Mol Sci 2021; 22:5123. [PMID: 34066087 PMCID: PMC8151504 DOI: 10.3390/ijms22105123] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Immune evasion is a key strategy adopted by tumor cells to escape the immune system while promoting their survival and metastatic spreading. Indeed, several mechanisms have been developed by tumors to inhibit immune responses. PD-1 is a cell surface inhibitory receptor, which plays a major physiological role in the maintenance of peripheral tolerance. In pathological conditions, activation of the PD-1/PD-Ls signaling pathway may block immune cell activation, a mechanism exploited by tumor cells to evade the antitumor immune control. Targeting the PD-1/PD-L1 axis has represented a major breakthrough in cancer treatment. Indeed, the success of PD-1 blockade immunotherapies represents an unprecedented success in the treatment of different cancer types. To improve the therapeutic efficacy, a deeper understanding of the mechanisms regulating PD-1 expression and signaling in the tumor context is required. We provide an overview of the current knowledge of PD-1 expression on both tumor-infiltrating T and NK cells, summarizing the recent evidence on the stimuli regulating its expression. We also highlight perspectives and limitations of the role of PD-L1 expression as a predictive marker, discuss well-established and novel potential approaches to improve patient selection and clinical outcome and summarize current indications for anti-PD1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Enrico Munari
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy;
| | - Francesca R. Mariotti
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Linda Quatrini
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Pietro Bertoglio
- Division of Thoracic Surgery, IRCCS Maggiore Teaching Hospital and Sant’Orsola University Hospital, 40133 Bologna, Italy;
| | - Nicola Tumino
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Paola Vacca
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Albino Eccher
- Pathology Unit, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Francesco Ciompi
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, 6543 SH Nijmegen, The Netherlands;
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (M.B.); (G.M.)
| | - Guido Martignoni
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (M.B.); (G.M.)
- Pathology Unit, Pederzoli Hospital, 37019 Peschiera del Garda, Italy
| | - Giuseppe Bogina
- Pathology Unit, IRCCS Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Lorenzo Moretta
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| |
Collapse
|
326
|
Ouyang T, Cao Y, Kan X, Chen L, Ren Y, Sun T, Yan L, Xiong B, Liang B, Zheng C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front Oncol 2021; 11:621639. [PMID: 34046338 PMCID: PMC8144509 DOI: 10.3389/fonc.2021.621639] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immune Checkpoint Inhibitors (ICI) have been progressively used in cancer treatment and produced unique toxicity profiles. This systematic review aims to comprehend the patterns and occurrence of treatment-related adverse events (trAEs) based on ICI. Methods PICOS/PRISMA methods were used to identify published English-language on PubMed, Web of Science, and Scopus from 2015 to 2020. Published clinical trials on ICI monotherapy, combined ICIs, and ICI plus other treatment with tabulated data on grade≥3 trAEs were included. Odds ratio (OR), χ2 tests were used to analyze for effect size and associations. Results This review included 145 clinical trials involving 21786 patients. Grade 3-5 trAEs were more common with ICI when they were plused with other treatments compared with ICI monotherapy(54.3% versus 17.7%, 46.1%, p<0.05). Grade 3-5 trAEs were also more common with CTLA-4 mAbs compared with anti-PD-1 and anti-PD-L1 (34.2% versus 15.1%, 13.6%, p<0.05). Hyperthyroidism (OR 3.8, 95%CI 1.7–8.6), nausea (OR 3.7, 95%CI 2.5–5.3), diarrhea (OR 2.7, 95%CI 2.2–3.2), colitis (OR 3.4, 95%CI 2.7–4.3), ALT increase (OR 4.9, 95%CI 3.9–6.1), AST increase (OR 3.8, 95%CI 3.0–4.9), pruritus (OR 2.4, 95%CI 1.5–3.9), rash (OR 2.8, 95%CI 2.1–3.8), fatigue (OR 2.8, 95%CI 2.2–3.7), decreased appetite (OR 2.4, 95%CI 1.5–3.8), and hypophysitis (OR 2.0, 95%CI 1.2–3.3) were more frequent with combined ICIs. Diarrhea (OR 8.1, 95%CI 6.4–10.3), colitis (OR 12.2, 95%CI 8.7–17.1), ALT increase (OR 5.1, 95%CI 3.5–7.4), AST increase (OR 4.2, 95%CI 2.8–6.3), pruritus (OR 4.1, 95%CI 2.0–8.4), rash (OR 4.4, 95%CI 2.9–6.8), hypophysitis (OR 12.1, 95%CI 6.3–23.4) were more common with CTLA-4 mAbs; whereas pneumonitis (OR 4.7, 95% CI 2.1–10.3) were more frequent with PD-1 mAbs. Conclusions Different immune checkpoint inhibitors are associated with different treatment-related adverse events profiles. A comprehensive data in this systematic review will provide comprehensive information for clinicians.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Liangliang Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
327
|
Pires da Silva I, Ahmed T, Reijers ILM, Weppler AM, Betof Warner A, Patrinely JR, Serra-Bellver P, Allayous C, Mangana J, Nguyen K, Zimmer L, Trojaniello C, Stout D, Lyle M, Klein O, Gerard CL, Michielin O, Haydon A, Ascierto PA, Carlino MS, Lebbe C, Lorigan P, Johnson DB, Sandhu S, Lo SN, Blank CU, Menzies AM, Long GV. Ipilimumab alone or ipilimumab plus anti-PD-1 therapy in patients with metastatic melanoma resistant to anti-PD-(L)1 monotherapy: a multicentre, retrospective, cohort study. Lancet Oncol 2021; 22:836-847. [PMID: 33989557 DOI: 10.1016/s1470-2045(21)00097-8] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Anti-PD-1 therapy (hereafter referred to as anti-PD-1) induces long-term disease control in approximately 30% of patients with metastatic melanoma; however, two-thirds of patients are resistant and will require further treatment. We aimed to determine the efficacy and safety of ipilimumab plus anti-PD-1 (pembrolizumab or nivolumab) compared with ipilimumab monotherapy in patients who are resistant to anti-PD-(L)1 therapy (hereafter referred to as anti-PD-[L]1). METHODS This multicentre, retrospective, cohort study, was done at 15 melanoma centres in Australia, Europe, and the USA. We included adult patients (aged ≥18 years) with metastatic melanoma (unresectable stage III and IV), who were resistant to anti-PD-(L)1 (innate or acquired resistance) and who then received either ipilimumab monotherapy or ipilimumab plus anti-PD-1 (pembrolizumab or nivolumab), based on availability of therapies or clinical factors determined by the physician, or both. Tumour response was assessed as per standard of care (CT or PET-CT scans every 3 months). The study endpoints were objective response rate, progression-free survival, overall survival, and safety of ipilimumab compared with ipilimumab plus anti-PD-1. FINDINGS We included 355 patients with metastatic melanoma, resistant to anti-PD-(L)1 (nivolumab, pembrolizumab, or atezolizumab), who had been treated with ipilimumab monotherapy (n=162 [46%]) or ipilimumab plus anti-PD-1 (n=193 [54%]) between Feb 1, 2011, and Feb 6, 2020. At a median follow-up of 22·1 months (IQR 9·5-30·9), the objective response rate was higher with ipilimumab plus anti-PD-1 (60 [31%] of 193 patients) than with ipilimumab monotherapy (21 [13%] of 162 patients; p<0·0001). Overall survival was longer in the ipilimumab plus anti-PD-1 group (median overall survival 20·4 months [95% CI 12·7-34·8]) than with ipilimumab monotherapy (8·8 months [6·1-11·3]; hazard ratio [HR] 0·50, 95% CI 0·38-0·66; p<0·0001). Progression-free survival was also longer with ipilimumab plus anti-PD-1 (median 3·0 months [95% CI 2·6-3·6]) than with ipilimumab (2·6 months [2·4-2·9]; HR 0·69, 95% CI 0·55-0·87; p=0·0019). Similar proportions of patients reported grade 3-5 adverse events in both groups (59 [31%] of 193 patients in the ipilimumab plus anti-PD-1 group vs 54 [33%] of 162 patients in the ipilimumab group). The most common grade 3-5 adverse events were diarrhoea or colitis (23 [12%] of 193 patients in the ipilimumab plus anti-PD-1 group vs 33 [20%] of 162 patients in the ipilimumab group) and increased alanine aminotransferase or aspartate aminotransferase (24 [12%] vs 15 [9%]). One death occurred with ipilimumab 26 days after the last treatment: a colon perforation due to immune-related pancolitis. INTERPRETATION In patients who are resistant to anti-PD-(L)1, ipilimumab plus anti-PD-1 seemed to yield higher efficacy than ipilimumab with a higher objective response rate, longer progression-free, and longer overall survival, with a similar rate of grade 3-5 toxicity. Ipilimumab plus anti-PD-1 should be favoured over ipilimumab alone as a second-line immunotherapy for these patients with advanced melanoma. FUNDING None.
Collapse
Affiliation(s)
- Ines Pires da Silva
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Westmead Hospital, Sydney, NSW, Australia; Blacktown Hospital, Sydney, NSW, Australia
| | - Tasnia Ahmed
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | | | - Alison M Weppler
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | | | | | | | - Clara Allayous
- AP-HP Dermatology, INSERM U976, Université de Paris, Saint Louis Hospital, Paris, France
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Khang Nguyen
- Westmead Hospital, Sydney, NSW, Australia; Blacktown Hospital, Sydney, NSW, Australia
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Claudia Trojaniello
- Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | - Dan Stout
- Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Megan Lyle
- Cairns Hospital, James Cook University, Cairns, QLD, Australia
| | - Oliver Klein
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
| | - Camille L Gerard
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Michielin
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Andrew Haydon
- Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Paolo A Ascierto
- Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | - Matteo S Carlino
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Westmead Hospital, Sydney, NSW, Australia; Blacktown Hospital, Sydney, NSW, Australia
| | - Celeste Lebbe
- AP-HP Dermatology, INSERM U976, Université de Paris, Saint Louis Hospital, Paris, France
| | - Paul Lorigan
- The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | | | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | | | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia.
| |
Collapse
|
328
|
Han D, van Akkooi ACJ, Straker RJ, Shannon AB, Karakousis GC, Wang L, Kim KB, Reintgen D. Current management of melanoma patients with nodal metastases. Clin Exp Metastasis 2021; 39:181-199. [PMID: 33961168 PMCID: PMC8102663 DOI: 10.1007/s10585-021-10099-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/22/2021] [Indexed: 12/26/2022]
Abstract
The management of melanoma patients with nodal metastases has undergone dramatic changes over the last decade. In the past, the standard of care for patients with a positive sentinel lymph node biopsy (SLNB) was a completion lymph node dissection (CLND), while patients with palpable macroscopic nodal disease underwent a therapeutic lymphadenectomy in cases with no evidence of systemic spread. However, studies have shown that SLN metastases present as a spectrum of disease, with certain SLN-based factors being prognostic of and correlated with outcomes. Furthermore, the results of key clinical trials demonstrate that CLND provides no survival benefit over nodal observation in positive SLN patients, while other clinical trials have shown that adjuvant immune checkpoint inhibitor therapy or targeted therapy after CLND is associated with a recurrence-free survival benefit. Given the efficacy of these systemic therapies in the adjuvant setting, these agents are now being evaluated and utilized as neoadjuvant treatments in patients with regionally-localized or resectable metastatic melanoma. Multiple options now exist to treat melanoma patients with nodal disease, and determining the best treatment course for a particular case requires an in-depth knowledge of current data and an informed discussion with the patient. This review will provide an overview of the various options for treating melanoma patients with nodal metastases and will discuss the data that supported the development of these treatment options.
Collapse
Affiliation(s)
- Dale Han
- Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code: L619, Portland, OR, 97239, USA.
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Richard J Straker
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Adrienne B Shannon
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Giorgos C Karakousis
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Lin Wang
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | - Kevin B Kim
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | - Douglas Reintgen
- Department of Surgery, Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
329
|
Pampena R, Michelini S, Lai M, Chester J, Pellacani G, Longo C. New systemic therapies for cutaneous melanoma: why, who and what. Ital J Dermatol Venerol 2021; 156:344-355. [PMID: 33913672 DOI: 10.23736/s2784-8671.21.06936-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Incidence of melanoma has been increasing in both sexes in the last decades. Advanced melanoma has always been one of the deadliest cancers worldwide due to his high metastatic capacity. In the last ten years, progresses in the knowledge of the molecular mechanisms involved in the melanoma development and progression, and in immune-response against melanoma, empowered the development of two new classes of systemic therapeutic agents: target-therapies and immunotherapies. Both classes consist of monoclonal antibodies inhibiting specific molecules. Target-therapies are selectively directed against cells harboring the BRAFV600-mutation, while immunotherapies target the two molecules involved in immune-checkpoint regulation, enhancing the immune response against the tumor: cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 receptor (PD-1). Target- and immunotherapy demonstrated to improve both progression-free and overall survival in melanoma patients either in metastatic or in adjuvant settings. Several drugs have been approved in recent years as monotherapy or in combination, and many other drugs are currently under investigation in clinical trials. In the current review on new systemic therapies for cutaneous melanoma, we revised the molecular basis and the mechanisms of actions of both target- and immunotherapy (why). We discussed who are the best candidate to receive such therapies in both the adjuvant and metastatic setting (who) and which were the most important efficacy and safety data on these drugs (what).
Collapse
Affiliation(s)
- Riccardo Pampena
- Centro Oncologico ad Alta Tecnologia Diagnostica, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Michela Lai
- Centro Oncologico ad Alta Tecnologia Diagnostica, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna Chester
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Pellacani
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Caterina Longo
- Centro Oncologico ad Alta Tecnologia Diagnostica, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy - .,Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
330
|
Bajaj S, Donnelly D, Call M, Johannet P, Moran U, Polsky D, Shapiro R, Berman R, Pavlick A, Weber J, Zhong J, Osman I. Melanoma Prognosis: Accuracy of the American Joint Committee on Cancer Staging Manual Eighth Edition. J Natl Cancer Inst 2021; 112:921-928. [PMID: 31977051 DOI: 10.1093/jnci/djaa008] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The American Joint Committee on Cancer (AJCC) maintains that the eighth edition of its Staging Manual (AJCC8) has improved accuracy compared with the seventh (AJCC7). However, there are concerns that implementation may disrupt analysis of active clinical trials for stage III patients. We used an independent cohort of melanoma patients to test the extent to which AJCC8 has improved prognostic accuracy compared with AJCC7. METHODS We analyzed a cohort of 1315 prospectively enrolled patients. We assessed primary tumor and nodal classification of stage I-III patients using AJCC7 and AJCC8 to assign disease stages at diagnosis. We compared recurrence-free (RFS) and overall survival (OS) using Kaplan-Meier curves and log-rank tests. We then compared concordance indices of discriminatory prognostic ability and area under the curve of 5-year survival to predict RFS and OS. All statistical tests were two-sided. RESULTS Stage IIC patients continued to have worse outcomes than stage IIIA patients, with a 5-year RFS of 26.5% (95% confidence interval [CI] = 12.8% to 55.1%) vs 56.0% (95% CI = 37.0% to 84.7%) by AJCC8 (P = .002). For stage I, removing mitotic index as a T classification factor decreased its prognostic value, although not statistically significantly (RFS concordance index [C-index] = 0.63, 95% CI = 0.56 to 0.69; to 0.56, 95% CI = 0.49 to 0.63, P = .07; OS C-index = 0.48, 95% CI = 0.38 to 0.58; to 0.48, 95% CI = 0.41 to 0.56, P = .90). For stage II, prognostication remained constant (RFS C-index = 0.65, 95% CI = 0.57 to 0.72; OS C-index = 0.61, 95% CI = 0.50 to 0.72), and for stage III, AJCC8 yielded statistically significantly enhanced prognostication for RFS (C-index = 0.65, 95% CI = 0.60 to 0.70; to 0.70, 95% CI = 0.66 to 0.75, P = .01). CONCLUSIONS Compared with AJCC7, we demonstrate that AJCC8 enables more accurate prognosis for patients with stage III melanoma. Restaging a large cohort of patients can enhance the analysis of active clinical trials.
Collapse
Affiliation(s)
- Shirin Bajaj
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Douglas Donnelly
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Melissa Call
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Paul Johannet
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Una Moran
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - David Polsky
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA.,Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Richard Shapiro
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Russell Berman
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Anna Pavlick
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Jeffrey Weber
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Judy Zhong
- Department of Population Health, NYU Langone School of Medicine, New York, NY, USA
| | - Iman Osman
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| |
Collapse
|
331
|
Khalifa J, Mazieres J, Gomez-Roca C, Ayyoub M, Moyal ECJ. Radiotherapy in the Era of Immunotherapy With a Focus on Non-Small-Cell Lung Cancer: Time to Revisit Ancient Dogmas? Front Oncol 2021; 11:662236. [PMID: 33968769 PMCID: PMC8097090 DOI: 10.3389/fonc.2021.662236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Radiation-induced immune effects have been extensively deciphered over the last few years, leading to the concept of the dual immune effect of radiotherapy with both immunostimulatory and immunosuppressive effects. This explains why radiotherapy alone is not able to drive a strong anti-tumor immune response in most cases, hence underlining the rationale for combining both radiotherapy and immunotherapy. This association has generated considerable interest and hundreds of trials are currently ongoing to assess such an association in oncology. However, while some trials have provided unprecedented results or shown much promise, many hopes have been dashed. Questions remain, therefore, as to how to optimize the combination of these treatment modalities. This narrative review aims at revisiting the old, well-established concepts of radiotherapy relating to dose, fractionation, target volumes and organs at risk in the era of immunotherapy. We then propose potential innovative approaches to be further assessed when considering a radio-immunotherapy association, especially in the field of non-small-cell lung cancer (NSCLC). We finally propose a framework to optimize the association, with pragmatic approaches depending on the stage of the disease.
Collapse
Affiliation(s)
- Jonathan Khalifa
- Department of Radiotherapy, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse – Oncopole, Toulouse, France
- Institut National de la Santé et de la Recherche Médicale U1037, Centre de Recherche contre le Cancer de Toulouse, Toulouse, France
| | - Julien Mazieres
- Department of Pulmonology, Centre Hospitalo-Universitaire Larrey, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - Carlos Gomez-Roca
- Institut National de la Santé et de la Recherche Médicale U1037, Centre de Recherche contre le Cancer de Toulouse, Toulouse, France
- Department of Medical Oncology, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse – Oncopole, Toulouse, France
| | - Maha Ayyoub
- Institut National de la Santé et de la Recherche Médicale U1037, Centre de Recherche contre le Cancer de Toulouse, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - Elizabeth Cohen-Jonathan Moyal
- Department of Radiotherapy, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse – Oncopole, Toulouse, France
- Institut National de la Santé et de la Recherche Médicale U1037, Centre de Recherche contre le Cancer de Toulouse, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
332
|
Berger DMS, Verver D, van der Noort V, Grünhagen DJ, Verhoef C, Al-Mamgani A, Zuur CL, van Akkooi ACJ, Balm AJM, Klop WMC. Therapeutic neck dissection in head and neck melanoma patients: Comparing extent of surgery and clinical outcome in two cohorts. Eur J Surg Oncol 2021; 47:2454-2459. [PMID: 33867173 DOI: 10.1016/j.ejso.2021.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/21/2021] [Accepted: 04/05/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The extent of surgical management of regional lymph nodes in the treatment of cutaneous head and neck melanoma on and anterior to O'Brien's watershed line is controversial. By comparing patients' cohorts of two separate melanoma expert centers we investigate the effectiveness of comprehensive versus (super-) selective neck dissection approach. METHODS Sixty patients with macroscopic (palpable) neck node metastases (N2b) from anterior scalp and face melanoma were retrospectively studied. Forty therapeutic modified radical neck dissections (MRND; levels I-V) combined with elective parotidectomy from The Netherlands Cancer Institute (NCI) were compared with 16 (super-) selective neck dissections [(S)SND; 3-4 levels] and 4 solely MRNDs from Erasmus Medical Center (EMC). Cohorts were analyzed for site of recurrence, overall survival (OS), melanoma-specific survival (MSS), and disease-free survival (DFS). RESULTS Clinical characteristics of patients were equal in both groups. In the NCI cohort 62.5% (n = 25) of patients recurred versus 65% (n = 13) in the EMC cohort. None of the NCI recurrences affected the parotid gland in contrast to 3 patients in the EMC group. Survival characteristics were not different between the two groups: OS (p = 0.56), MSS (p = 0.98), DFS (p = 0.92). CONCLUSION This study does not support to continue the practice of routine elective parotidectomy and MRND in melanoma patients undergoing a lymph node dissection for macroscopic (palpable) nodal disease and justifies (S)SND.
Collapse
Affiliation(s)
- Danique M S Berger
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands.
| | - Danielle Verver
- Department of Surgical Oncology, Erasmus MC-Cancer Institute, Rotterdam, the Netherlands
| | - Vincent van der Noort
- Department of Epidemiology and Biostatistics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology, Erasmus MC-Cancer Institute, Rotterdam, the Netherlands
| | - Cees Verhoef
- Department of Surgical Oncology, Erasmus MC-Cancer Institute, Rotterdam, the Netherlands
| | - Abrahim Al-Mamgani
- Department of Radiotherapy, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Charlotte L Zuur
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Maxillofacial Surgery, Academic Medical Center, Amsterdam, the Netherlands
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Alfons J M Balm
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Maxillofacial Surgery, Academic Medical Center, Amsterdam, the Netherlands
| | - W Martin C Klop
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Maxillofacial Surgery, Academic Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
333
|
Boulva K, Apte S, Yu A, Tran A, Shorr R, Song X, Ong M, Nessim C. Contemporary Neoadjuvant Therapies for High-Risk Melanoma: A Systematic Review. Cancers (Basel) 2021; 13:1905. [PMID: 33920967 PMCID: PMC8071293 DOI: 10.3390/cancers13081905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 11/16/2022] Open
Abstract
Despite advances in adjuvant immuno- and targeted therapies, the risk of relapse for stage III melanoma remains high. With 43 active entries on clinicaltrials.gov (8 July 2020), there is a surge of interest in the role of contemporary therapies in the neoadjuvant setting. We conducted a systematic review of trials performed in the last decade evaluating neoadjuvant targeted, immuno- or intralesional therapy for resectable stage III or IV melanoma. Database searches of Medline, Embase, and the Cochrane Central Register of Controlled Trials were conducted from inception to 13 February 2020. Two reviewers assessed titles, abstracts, and full texts. Trials investigating contemporary neoadjuvant therapies in high-risk melanoma were included. Eight phase II trials (4 randomized and 4 single-arm) involving 450 patients reported on neoadjuvant anti-BRAF/MEK targeted therapy (3), anti-PD-1/CTLA-4 immunotherapy (3), and intralesional therapy (2). The safest and most efficacious regimens were dabrafenib/trametinib and combination ipilimumab (1 mg/kg) + nivolumab (3 mg/kg). Pathologic complete response (pCR) and adverse events were comparable. Ipilimumab + nivolumab exhibited longer RFS. Contemporary neoadjuvant therapies are not only safe, but also demonstrate remarkable pCR and RFS-outcomes which are regarded as meaningful surrogates for long-term survival. Studies defining predictors of pCR, its correlation with oncologic outcomes, and phase III trials comparing neoadjuvant therapy to standard of care will be crucial.
Collapse
Affiliation(s)
- Kerianne Boulva
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
| | - Sameer Apte
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| | - Ashley Yu
- Department of Family Medicine, McMaster University, Hamilton, ON L8P 1H6, Canada;
| | - Alexandre Tran
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| | - Risa Shorr
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| | - Xinni Song
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
- Division of Medical Oncology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Michael Ong
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
- Division of Medical Oncology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Carolyn Nessim
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| |
Collapse
|
334
|
Sahovaler A, Ziai H, Cardemil F, Huang SH, Su J, Goldstein DP, Gilbert R, Hosni A, Hope A, Waldron J, Spreafico A, Monteiro E, Witterick I, Irish J, Gullane P, Xu W, O'Sullivan B, de Almeida JR. Importance of Margins, Radiotherapy, and Systemic Therapy in Mucosal Melanoma of the Head and Neck. Laryngoscope 2021; 131:2269-2276. [PMID: 33856051 DOI: 10.1002/lary.29555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES/HYPOTHESIS The ideal strategy in the treatment of mucosal melanoma of the head and neck (MMHN) remains unclear. Our objective was to evaluate the importance of surgical margins, radiotherapy, and systemic therapy in MMHN. STUDY DESIGN Retrospective Single Institutional Review. METHODS Retrospective review of patients with MMHN treated at a tertiary care oncology center between 1999 and 2016. RESULTS Seventy-six patients were included, 60 of whom were treated with curative intent. Negative or close margins compared with positive margins were associated with higher 3-year overall survival (OS) (62% vs. 29% vs. 13% P = .012), disease-free survival (33% vs. 29% vs. 4% P = .003), and distant control (48% vs. 29% vs. 22% P = .039). Cases with pre-/postoperative radiotherapy had a marginally higher locoregional control versus without (69% vs. 59%, P = .117). Immunotherapy for recurrent and/or metastatic disease was associated with an increase in 3-year OS (15% vs. 3% P = .01). CONCLUSION Achieving negative surgical margins is relevant in disease control. Despite small sample size, our data suggest that radiotherapy may enhance surgical outcomes. Immunotherapy has therapeutic benefit. LEVEL OF EVIDENCE 3 Laryngoscope, 2021.
Collapse
Affiliation(s)
- Axel Sahovaler
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hedyeh Ziai
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Felipe Cardemil
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Shao Hui Huang
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jie Su
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ralph Gilbert
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ali Hosni
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - John Waldron
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Eric Monteiro
- Department of Otolaryngology-Head and Neck Surgery, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Ian Witterick
- Department of Otolaryngology-Head and Neck Surgery, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Irish
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Gullane
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Brian O'Sullivan
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - John R de Almeida
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
335
|
Yu CI, Martinek J, Wu TC, Kim KI, George J, Ahmadzadeh E, Maser R, Marches F, Metang P, Authie P, Oliveira VKP, Wang VG, Chuang JH, Robson P, Banchereau J, Palucka K. Human KIT+ myeloid cells facilitate visceral metastasis by melanoma. J Exp Med 2021; 218:211995. [PMID: 33857287 PMCID: PMC8056753 DOI: 10.1084/jem.20182163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/30/2020] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Metastasis of melanoma significantly worsens prognosis; thus, therapeutic interventions that prevent metastasis could improve patient outcomes. Here, we show using humanized mice that colonization of distant visceral organs with melanoma is dependent upon a human CD33+CD11b+CD117+ progenitor cell subset comprising <4% of the human CD45+ leukocytes. Metastatic tumor-infiltrating CD33+ cells from patients and humanized (h)NSG-SGM3 mice showed converging transcriptional profiles. Single-cell RNA-seq analysis identified a gene signature of a KIT/CD117-expressing CD33+ subset that correlated with decreased overall survival in a TCGA melanoma cohort. Thus, human CD33+CD11b+CD117+ myeloid cells represent a novel candidate biomarker as well as a therapeutic target for metastatic melanoma.
Collapse
Affiliation(s)
- Chun I Yu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT.,The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| | - Jan Martinek
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Te-Chia Wu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Kyung In Kim
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | | | - Rick Maser
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| | | | - Patrick Metang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| | - Pierre Authie
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| | | | - Victor G Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT
| | - Jacques Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT.,The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT.,The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
336
|
Ghisoni E, Wicky A, Bouchaab H, Imbimbo M, Delyon J, Gautron Moura B, Gérard CL, Latifyan S, Özdemir BC, Caikovski M, Pradervand S, Tavazzi E, Gatta R, Marandino L, Valabrega G, Aglietta M, Obeid M, Homicsko K, Mederos Alfonso NN, Zimmermann S, Coukos G, Peters S, Cuendet MA, Di Maio M, Michielin O. Late-onset and long-lasting immune-related adverse events from immune checkpoint-inhibitors: An overlooked aspect in immunotherapy. Eur J Cancer 2021; 149:153-164. [PMID: 33865201 DOI: 10.1016/j.ejca.2021.03.010] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionised cancer therapy but frequently cause immune-related adverse events (irAEs). Description of late-onset and duration of irAEs in the literature is often incomplete. METHODS To investigate reporting and incidence of late-onset and long-lasting irAEs, we reviewed all registration trials leading to ICI's approval by the US FDA and/or EMA up to December 2019. We analysed real-world data from all lung cancer (LC) and melanoma (Mel) patients treated with approved ICIs at the University Hospital of Lausanne (CHUV) from 2011 to 2019. To account for the immortal time bias, we used a time-dependent analysis to assess the potential association between irAEs and overall survival (OS). RESULTS Duration of irAEs and proportion of patients with ongoing toxicities at data cut-off were not specified in 56/62 (90%) publications of ICIs registration trials. In our real-world analysis, including 437 patients (217 LC, 220 Mel), 229 (52.4%) experienced at least one grade ≥2 toxicity, for a total of 318 reported irAEs, of which 112 (35.2%) were long-lasting (≥6 months) and about 40% were ongoing at a median follow-up of 369 days [194-695] or patient death. The cumulative probability of irAE onset from treatment initiation was 42.8%, 51.0% and 57.3% at 6, 12 and 24 months, respectively. The rate of ongoing toxicity from the time of first toxicity onset was 42.8%, 38.4% and 35.7% at 6, 12 and 24 months. Time-dependent analysis showed no significant association between the incidence of irAEs and OS in both cohorts (log Rank p = 0.67 and 0.19 for LC and Mel, respectively). CONCLUSIONS Late-onset and long-lasting irAEs are underreported but common events during ICIs therapy. Time-dependent survival analysis is advocated to assess their impact on OS. Real-world evidence is warranted to fully capture and characterise late-onset and long-lasting irAEs in order to implement appropriate strategies for patient surveillance and follow-up.
Collapse
Affiliation(s)
- E Ghisoni
- Department of Oncology, Lausanne University Hospital, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - A Wicky
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - H Bouchaab
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - M Imbimbo
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - J Delyon
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - B Gautron Moura
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - C L Gérard
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - S Latifyan
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - B C Özdemir
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - M Caikovski
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - S Pradervand
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - E Tavazzi
- Department of Information Engineering, University of Padova, Italy
| | - R Gatta
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - L Marandino
- Department of Oncology, University of Torino, Italy
| | - G Valabrega
- Department of Oncology, University of Torino, Italy; Candiolo Cancer Institute, FPO, IRCCS, Candiolo (TO), Italy
| | - M Aglietta
- Department of Oncology, University of Torino, Italy; Candiolo Cancer Institute, FPO, IRCCS, Candiolo (TO), Italy
| | - M Obeid
- Service Immunologie et Allergie, Lausanne University Hospital, Switzerland
| | - K Homicsko
- Department of Oncology, Lausanne University Hospital, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | | | - S Zimmermann
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - G Coukos
- Department of Oncology, Lausanne University Hospital, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - S Peters
- Department of Oncology, Lausanne University Hospital, Switzerland
| | - M A Cuendet
- Department of Oncology, Lausanne University Hospital, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, USA
| | - M Di Maio
- Department of Oncology, University of Torino, Italy; Medical Oncology, A.O. Ordine Mauriziano, Torino, Italy
| | - O Michielin
- Department of Oncology, Lausanne University Hospital, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland.
| |
Collapse
|
337
|
Eggermont AMM, Blank CU, Mandalà M, Long GV, Atkinson VG, Dalle S, Haydon AM, Meshcheryakov A, Khattak A, Carlino MS, Sandhu S, Larkin J, Puig S, Ascierto PA, Rutkowski P, Schadendorf D, Koornstra R, Hernandez-Aya L, Di Giacomo AM, van den Eertwegh AJM, Grob JJ, Gutzmer R, Jamal R, Lorigan PC, van Akkooi ACJ, Krepler C, Ibrahim N, Marreaud S, Kicinski M, Suciu S, Robert C. Adjuvant pembrolizumab versus placebo in resected stage III melanoma (EORTC 1325-MG/KEYNOTE-054): distant metastasis-free survival results from a double-blind, randomised, controlled, phase 3 trial. Lancet Oncol 2021; 22:643-654. [PMID: 33857412 DOI: 10.1016/s1470-2045(21)00065-6] [Citation(s) in RCA: 257] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The European Organisation for Research and Treatment of Cancer (EORTC) 1325/KEYNOTE-054 trial assessed pembrolizumab versus placebo in patients with resected high-risk stage III melanoma. At 15-month median follow-up, pembrolizumab improved recurrence-free survival (hazard ratio [HR] 0·57 [98·4% CI 0·43-0·74], p<0·0001) compared with placebo, leading to its approval in the USA and Europe. This report provides the final results for the secondary efficacy endpoint, distant metastasis-free survival and an update of the recurrence-free survival results. METHODS This double-blind, randomised, controlled, phase 3 trial was done at 123 academic centres and community hospitals across 23 countries. Patients aged 18 years or older with complete resection of cutaneous melanoma metastatic to lymph node, classified as American Joint Committee on Cancer staging system, seventh edition (AJCC-7) stage IIIA (at least one lymph node metastasis >1 mm), IIIB, or IIIC (without in-transit metastasis), and with an Eastern Cooperative Oncology Group performance status of 0 or 1 were eligible. Patients were randomly assigned (1:1) via a central interactive voice response system to receive intravenous pembrolizumab 200 mg or placebo every 3 weeks for up to 18 doses or until disease recurrence or unacceptable toxicity. Randomisation was stratified according to disease stage and region, using a minimisation technique, and clinical investigators, patients, and those collecting or analysing the data were masked to treatment assignment. The two coprimary endpoints were recurrence-free survival in the intention-to-treat (ITT) population and in patients with PD-L1-positive tumours. The secondary endpoint reported here was distant metastasis-free survival in the ITT and PD-L1-positive populations. This study is registered with ClinicalTrials.gov, NCT02362594, and EudraCT, 2014-004944-37. FINDINGS Between Aug 26, 2015, and Nov 14, 2016, 1019 patients were assigned to receive either pembrolizumab (n=514) or placebo (n=505). At an overall median follow-up of 42·3 months (IQR 40·5-45·9), 3·5-year distant metastasis-free survival was higher in the pembrolizumab group than in the placebo group in the ITT population (65·3% [95% CI 60·9-69·5] in the pembrolizumab group vs 49·4% [44·8-53·8] in the placebo group; HR 0·60 [95% CI 0·49-0·73]; p<0·0001). In the 853 patients with PD-L1-positive tumours, 3·5-year distant metastasis-free survival was 66·7% (95% CI 61·8-71·2) in the pembrolizumab group and 51·6% (46·6-56·4) in the placebo group (HR 0·61 [95% CI 0·49-0·76]; p<0·0001). Recurrence-free survival remained longer in the pembrolizumab group 59·8% (95% CI 55·3-64·1) than the placebo group 41·4% (37·0-45·8) at this 3·5-year follow-up in the ITT population (HR 0·59 [95% CI 0·49-0·70]) and in those with PD-L1-positive tumours 61·4% (56·3-66·1) in the pembrolizumab group and 44·1% (39·2-48·8) in the placebo group (HR 0·59 [95% CI 0·49-0·73]). INTERPRETATION Pembrolizumab adjuvant therapy provided a significant and clinically meaningful improvement in distant metastasis-free survival at a 3·5-year median follow-up, which was consistent with the improvement in recurrence-free survival. Therefore, the results of this trial support the indication to use adjuvant pembrolizumab therapy in patients with resected high risk stage III cutaneous melanoma. FUNDING Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Princess Máxima Center, Utrecht, Netherlands; University Medical Center Utrecht, Utrecht, Netherlands.
| | - Christian U Blank
- Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Mario Mandalà
- Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Georgina V Long
- Melanoma Institute Australia, the University of Sydney, and Mater and Royal North Shore Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Adnan Khattak
- Fiona Stanley Hospital & Edith Cowan University, Perth, WA, Australia
| | - Matteo S Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia and the University of Sydney, Sydney, NSW, Australia
| | | | | | - Susana Puig
- Hospital Clinic de Barcelona, Universitat de Barcelona, Spain & Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS "Fondazione G Pascale", Naples, Italy
| | - Piotr Rutkowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Dirk Schadendorf
- University Hospital Essen, Essen and German Cancer Consortium, Heidelberg, Germany
| | - Rutger Koornstra
- Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | | | | | | | | | - Ralf Gutzmer
- Skin Cancer Center, Hannover Medical School, Hannover, Germany
| | - Rahima Jamal
- Centre Hospitalier de l'Université de Montréal (CHUM), Centre de recherche du CHUM, Montreal, QC, Canada
| | | | | | | | | | | | | | | | - Caroline Robert
- Gustave Roussy and Paris-Saclay University, Villejuif, France
| | | |
Collapse
|
338
|
Adjuvant pembrolizumab versus placebo in resected stage III melanoma (EORTC 1325-MG/KEYNOTE-054): health-related quality-of-life results from a double-blind, randomised, controlled, phase 3 trial. Lancet Oncol 2021; 22:655-664. [PMID: 33857414 DOI: 10.1016/s1470-2045(21)00081-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/04/2021] [Accepted: 02/04/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND The European Organisation for Research and Treatment of Cancer (EORTC) 1325-MG/KEYNOTE-054 trial in patients with resected, high-risk stage III melanoma demonstrated improved recurrence-free survival with adjuvant pembrolizumab compared with placebo (hazard ratio 0·57 [98·4% CI 0·43-0·74]; p<0·0001). This study reports the results from the health-related quality-of-life (HRQOL) exploratory endpoint. METHODS This double-blind, randomised, controlled, phase 3 trial was done at 123 academic centres and community hospitals across 23 countries. Patients aged 18 years or older with previously untreated histologically confirmed stage IIIA, IIIB, or IIIC resected cutaneous melanoma, and an Eastern Cooperative Oncology Group performance status score of 1 or 0 were eligible. Patients were randomly assigned (1:1) using a central interactive voice-response system on the basis of a minimisation technique stratified for stage and geographic region to receive intravenously 200 mg pembrolizumab or placebo. Treatment was administered every 3 weeks for 1 year, or until disease recurrence, unacceptable toxicity, or death. The primary endpoint of the trial was recurrence-free survival (reported elsewhere). HRQOL was a prespecified exploratory endpoint, with global health/quality of life (GHQ) over 2 years measured by the EORTC QLQ-C30 as the primary analysis. Analyses were done in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT02362594, and EudraCT, 2014-004944-37, and long-term follow-up is ongoing. FINDINGS Between Aug 26, 2015, and Nov 14, 2016, 1019 patients were assigned to pembrolizumab (n=514) or placebo (n=505). Median follow-up was 15·1 months (IQR 12·8-16·9) at the time of this analysis. HRQOL compliance was greater than 90% at baseline, greater than 70% during the first year, and greater than 60% thereafter for both groups. Because of low absolute compliance numbers at later follow-up, the analysis was truncated to week 84. Baseline GHQ scores were similar between groups (77·55 [SD 18·20] in the pembrolizumab group and 76·54 [17·81] in the placebo group) and remained stable over time. The difference in average GHQ score between the two groups over the 2 years was -2·2 points (95% CI -4·3 to -0·2). The difference in average score during treatment was -1·1 points (95% CI -3·2 to 0·9) and the difference in average score after treatment was -2·2 points (-4·8 to 0·4). These differences are within the 5-point clinical relevance threshold for the QLQ-C30 and are therefore clinically non-significant. INTERPRETATION Pembrolizumab does not result in a clinically significant decrease in HRQOL compared with placebo when given as adjuvant therapy for patients with resected, high-risk stage III melanoma. These results support the use of adjuvant pembrolizumab in this setting. FUNDING Merck Sharp & Dohme.
Collapse
|
339
|
Broman KK, Hughes T, Dossett L, Sun J, Kirichenko D, Carr MJ, Sharma A, Bartlett EK, Nijhuis AAG, Thompson JF, Hieken TJ, Kottschade L, Downs J, Gyorki DE, Stahlie E, van Akkooi A, Ollila DW, Frank J, Song Y, Karakousis G, Moncrieff M, Nobes J, Vetto J, Han D, Farma JM, Deneve JL, Fleming MD, Perez MC, Lowe MC, Olofsson Bagge R, Mattsson J, Lee AY, Berman RS, Chai H, Kroon HM, Teras J, Teras RM, Farrow NE, Beasley G, Hui JYC, Been L, Kruijff S, Kim Y, Naqvi SMH, Sarnaik AA, Sondak VK, Zager JS. Active surveillance of patients who have sentinel node positive melanoma: An international, multi-institution evaluation of adoption and early outcomes after the Multicenter Selective Lymphadenectomy Trial II (MSLT-2). Cancer 2021; 127:2251-2261. [PMID: 33826754 DOI: 10.1002/cncr.33483] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND For patients with sentinel lymph node (SLN)-positive cutaneous melanoma, the Second Multicenter Selective Lymphadenectomy trial demonstrated equivalent disease-specific survival (DSS) with active surveillance using nodal ultrasound versus completion lymph node dissection (CLND). Adoption and outcomes of active surveillance in clinical practice and in adjuvant therapy recipients are unknown. METHODS In a retrospective cohort of SLN-positive adults treated at 21 institutions in Australia, Europe, and the United States from June 2017 to November 2019, the authors evaluated the impact of active surveillance and adjuvant therapy on all-site recurrence-free survival (RFS), isolated nodal RFS, distant metastasis-free survival (DMFS), and DSS using Kaplan-Meier curves and Cox proportional hazard models. RESULTS Among 6347 SLN biopsies, 1154 (18%) were positive and had initial negative distant staging. In total, 965 patients (84%) received active surveillance, 189 (16%) underwent CLND. Four hundred thirty-nine patients received adjuvant therapy (surveillance, 38%; CLND, 39%), with the majority (83%) receiving anti-PD-1 immunotherapy. After a median follow-up of 11 months, 220 patients developed recurrent disease (surveillance, 19%; CLND, 22%), and 24 died of melanoma (surveillance, 2%; CLND, 4%). Sixty-eight patients had an isolated nodal recurrence (surveillance, 6%; CLND, 4%). In patients who received adjuvant treatment without undergoing prior CLND, all isolated nodal recurrences were resectable. On risk-adjusted multivariable analyses, CLND was associated with improved isolated nodal RFS (hazard ratio [HR], 0.36; 95% CI, 0.15-0.88), but not all-site RFS (HR, 0.68; 95% CI, 0.45-1.02). Adjuvant therapy improved all-site RFS (HR, 0.52; 95% CI, 0.47-0.57). DSS and DMFS did not differ by nodal management or adjuvant treatment. CONCLUSIONS Active surveillance has been adopted for most SLN-positive patients. At initial assessment, real-world outcomes align with randomized trial findings, including in adjuvant therapy recipients. LAY SUMMARY For patients with melanoma of the skin and microscopic spread to lymph nodes, monitoring with ultrasound is an alternative to surgically removing the remaining lymph nodes. The authors studied adoption and real-world outcomes of ultrasound monitoring in over 1000 patients treated at 21 centers worldwide, finding that most patients now have ultrasounds instead of surgery. Although slightly more patients have cancer return in the lymph nodes with this strategy, typically, it can be removed with delayed surgery. Compared with up-front surgery, ultrasound monitoring results in the same overall risk of melanoma coming back at any location or of dying from melanoma.
Collapse
Affiliation(s)
- Kristy Kummerow Broman
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, Florida
| | - Tasha Hughes
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Lesly Dossett
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - James Sun
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Dennis Kirichenko
- Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, Florida
| | - Michael J Carr
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Avinash Sharma
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Edmund K Bartlett
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amanda A G Nijhuis
- Department of Surgery, Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - John F Thompson
- Department of Surgery, Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Tina J Hieken
- Department of Surgery, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Lisa Kottschade
- Department of Surgery, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jennifer Downs
- Division of Cancer Surgery, Peter MacCallum Cancer Center, Melbourne, Australia
| | - David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Emma Stahlie
- Division of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alexander van Akkooi
- Division of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - David W Ollila
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Jill Frank
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Yun Song
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Giorgos Karakousis
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marc Moncrieff
- Department of Plastic Surgery, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Jenny Nobes
- Department of Plastic Surgery, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - John Vetto
- Department of Surgery, Oregon Health & Science University, Portland, Oregon
| | - Dale Han
- Department of Surgery, Oregon Health & Science University, Portland, Oregon
| | - Jeffrey M Farma
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Martin D Fleming
- Department of Surgery, University of Tennessee, Memphis, Tennessee
| | | | - Michael C Lowe
- Department of Surgery, Emory University, Atlanta, Georgia
| | - Roger Olofsson Bagge
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Mattsson
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ann Y Lee
- Department of Surgery, NYU Langone Health, New York, New York
| | | | - Harvey Chai
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
| | - Hidde M Kroon
- Discipline of Surgery, Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
| | - Juri Teras
- Surgery Clinic, North Estonia Medical Center Foundation, Tallinn, Estonia
| | - Roland M Teras
- Surgery Clinic, North Estonia Medical Center Foundation, Tallinn, Estonia
| | - Norma E Farrow
- Department of Surgery, Duke University, Durham, North Carolina
| | - Georgia Beasley
- Department of Surgery, Duke University, Durham, North Carolina
| | | | - Lukas Been
- Department of Surgical Oncology, University Medical Center, Groningen, the Netherlands
| | - Schelto Kruijff
- Department of Surgical Oncology, University Medical Center, Groningen, the Netherlands
| | - Youngchul Kim
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | | | - Amod A Sarnaik
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, Florida
| | - Vernon K Sondak
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, Florida
| | - Jonathan S Zager
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, Florida
| |
Collapse
|
340
|
Chhabra N, Kennedy J. A Review of Cancer Immunotherapy Toxicity: Immune Checkpoint Inhibitors. J Med Toxicol 2021; 17:411-424. [PMID: 33826117 DOI: 10.1007/s13181-021-00833-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy, which leverages features of the immune system to target neoplastic cells, has revolutionized the treatment of cancer. The use of these therapies has rapidly expanded in the past two decades. Immune checkpoint inhibitors represent one drug class within immunotherapy with its first agent FDA-approved in 2011. Immune checkpoint inhibitors act by disrupting inhibitory signals from neoplastic cells to immune effector cells, allowing activated T-cells to target these neoplastic cells. Unique adverse effects associated with immune checkpoint inhibitors are termed immune-related adverse effects (irAEs) and are usually immunostimulatory in nature. Almost all organ systems may be affected by irAEs including the dermatologic, gastrointestinal, pulmonary, endocrine, and cardiovascular systems. These effects range from mild to life-threatening, and their onset can be delayed several weeks or months. For mild irAEs, symptomatic care is usually sufficient. For higher grade irAEs, discontinuation of therapy and initiation of immunosuppressive therapy may be necessary. The management of patients with irAEs involves multidisciplinary care coordination with respect to the long-term goals the individual patient. Clinicians must be aware of the unique and sometimes fatal toxicologic profiles associated with immunotherapies to ensure prompt diagnosis and appropriate management.
Collapse
Affiliation(s)
- Neeraj Chhabra
- Cook County Health, Department of Emergency Medicine, Division of Medical Toxicology, 1950 W Polk Street, 7th Floor, Chicago, IL, 60612, USA. .,Toxikon Consortium, Chicago, IL, USA.
| | - Joseph Kennedy
- Cook County Health, Department of Emergency Medicine, Division of Medical Toxicology, 1950 W Polk Street, 7th Floor, Chicago, IL, 60612, USA.,Toxikon Consortium, Chicago, IL, USA
| |
Collapse
|
341
|
Cytoreduction and the Optimization Of Immune Checkpoint Inhibition with Radiation Therapy. Int J Radiat Oncol Biol Phys 2021; 108:17-26. [PMID: 32819613 DOI: 10.1016/j.ijrobp.2019.12.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/10/2019] [Accepted: 12/24/2019] [Indexed: 12/29/2022]
|
342
|
Massagué J, Ganesh K. Metastasis-Initiating Cells and Ecosystems. Cancer Discov 2021; 11:971-994. [PMID: 33811127 PMCID: PMC8030695 DOI: 10.1158/2159-8290.cd-21-0010] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 11/16/2022]
Abstract
Metastasis is initiated and sustained through therapy by cancer cells with stem-like and immune-evasive properties, termed metastasis-initiating cells (MIC). Recent progress suggests that MICs result from the adoption of a normal regenerative progenitor phenotype by malignant cells, a phenotype with intrinsic programs to survive the stresses of the metastatic process, undergo epithelial-mesenchymal transitions, enter slow-cycling states for dormancy, evade immune surveillance, establish supportive interactions with organ-specific niches, and co-opt systemic factors for growth and recurrence after therapy. Mechanistic understanding of the molecular mediators of MIC phenotypes and host tissue ecosystems could yield cancer therapeutics to improve patient outcomes. SIGNIFICANCE: Understanding the origins, traits, and vulnerabilities of progenitor cancer cells with the capacity to initiate metastasis in distant organs, and the host microenvironments that support the ability of these cells to evade immune surveillance and regenerate the tumor, is critical for developing strategies to improve the prevention and treatment of advanced cancer. Leveraging recent progress in our understanding of the metastatic process, here we review the nature of MICs and their ecosystems and offer a perspective on how this knowledge is informing innovative treatments of metastatic cancers.
Collapse
Affiliation(s)
- Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, New York.
| | - Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York.
- Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
343
|
Poizeau F, Kerbrat S, Happe A, Rault C, Drezen E, Balusson F, Tuppin P, Guillot B, Thuret A, Boussemart L, Dinulescu M, Pracht M, Lesimple T, Droitcourt C, Oger E, Dupuy A. Patients with Metastatic Melanoma Receiving Anticancer Drugs: Changes in Overall Survival, 2010–2017. J Invest Dermatol 2021; 141:830-839.e3. [DOI: 10.1016/j.jid.2020.07.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022]
|
344
|
Akintola OO, Reardon DA. The Current Landscape of Immune Checkpoint Blockade in Glioblastoma. Neurosurg Clin N Am 2021; 32:235-248. [PMID: 33781505 DOI: 10.1016/j.nec.2020.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The glioblastoma tumor microenvironment is highly immunosuppressed. This immunosuppressive state is engineered by inhibitory molecules secreted by tumor cells that limit activation of immune effector cells, drive T-cell exhaustion, and enhance the immunosuppressive action of tumor-associated myeloid cells. Immunotherapeutic approaches have sought to combat glioblastoma microenvironment immunosuppression with agents such as immune checkpoint inhibitors. Although immune checkpoint blockade in glioblastoma has yielded disappointing results thus far, there is significant interest in the combination of immune checkpoint blockade with other approaches to enhance response.
Collapse
Affiliation(s)
- Oluwatosin O Akintola
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Massachusetts General Hospital Cancer Center, 450 Brookline Avenue, Boston, MA 02215-5450, USA.
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| |
Collapse
|
345
|
Ottaviano M, Giunta EF, Tortora M, Curvietto M, Attademo L, Bosso D, Cardalesi C, Rosanova M, De Placido P, Pietroluongo E, Riccio V, Mucci B, Parola S, Vitale MG, Palmieri G, Daniele B, Simeone E, on behalf of SCITO YOUTH. BRAF Gene and Melanoma: Back to the Future. Int J Mol Sci 2021; 22:ijms22073474. [PMID: 33801689 PMCID: PMC8037827 DOI: 10.3390/ijms22073474] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
As widely acknowledged, 40-50% of all melanoma patients harbour an activating BRAF mutation (mostly BRAF V600E). The identification of the RAS-RAF-MEK-ERK (MAP kinase) signalling pathway and its targeting has represented a valuable milestone for the advanced and, more recently, for the completely resected stage III and IV melanoma therapy management. However, despite progress in BRAF-mutant melanoma treatment, the two different approaches approved so far for metastatic disease, immunotherapy and BRAF+MEK inhibitors, allow a 5-year survival of no more than 60%, and most patients relapse during treatment due to acquired mechanisms of resistance. Deep insight into BRAF gene biology is fundamental to describe the acquired resistance mechanisms (primary and secondary) and to understand the molecular pathways that are now being investigated in preclinical and clinical studies with the aim of improving outcomes in BRAF-mutant patients.
Collapse
Affiliation(s)
- Margaret Ottaviano
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
- Correspondence:
| | - Emilio Francesco Giunta
- Department of Precision Medicine, Università Degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | - Marianna Tortora
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
| | - Marcello Curvietto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; (M.C.); (M.G.V.); (E.S.)
| | - Laura Attademo
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Davide Bosso
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Cinzia Cardalesi
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Mario Rosanova
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Vittorio Riccio
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Brigitta Mucci
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Sara Parola
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Maria Grazia Vitale
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; (M.C.); (M.G.V.); (E.S.)
| | - Giovannella Palmieri
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
| | - Bruno Daniele
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Ester Simeone
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; (M.C.); (M.G.V.); (E.S.)
| | | |
Collapse
|
346
|
Abstract
The management of melanoma significantly improved within the last 25 years. Chemotherapy was the first approved systemic therapeutic approach and resulted in a median overall of survival less than 1 year, without survival improvement in phase III trials. High-dose interferon α2b and IL-2 were introduced for resectable high-risk and advanced disease, respectively, resulting in improved survival and response rates. The anti-CTLA4 and anti-programmed death 1 monoclonal antibodies along with BRAF/MEK targeted therapies are the dominant therapeutic classes of agent for melanoma. This article provides an historic overview of the evolution of melanoma management.
Collapse
|
347
|
Tarhini AA. Adjuvant Therapy of Melanoma. Hematol Oncol Clin North Am 2021; 35:73-84. [PMID: 33759774 DOI: 10.1016/j.hoc.2020.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
as adjuvant therapy for high-risk melanoma was extensively studied in regimens that varied by dosage, route of administration, formulation, and therapy duration. The high-dose regimen (HDI) showed significant improvements in relapse-free survival (RFS) in 3 trials and overall survival (OS) in 2. Ipilimumab at 3 mg/kg demonstrated significant OS benefits compared with HDI and less toxicity compared with ipilimumab at 10 mg/kg. More recently, the standard of care has changed in favor of nivolumab and pembrolizumab and BRAF-MEK inhibitors dabrafenib plus trametinib (for BRAF mutated melanoma), based on significant RFS benefits and more favorable toxicity profiles.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 10920 McKinley Drive, Tampa, FL 33612, USA; Department of Immunology, Moffitt Cancer Center and Research Institute, 10902 USF Magnolia Drive, Tampa, FL 33612, USA.
| |
Collapse
|
348
|
Zam W, Ali L. Immune checkpoint inhibitors in the treatment of cancer. ACTA ACUST UNITED AC 2021; 17:103-113. [PMID: 33823768 DOI: 10.2174/1574884716666210325095022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Immunotherapy drugs, known as immune checkpoint inhibitors (ICIs), work by blocking checkpoint proteins from binding with their partner proteins. The two main pathways that are specifically targeted in clinical practice are cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) that showed potent immune-modulatory effects through their function as negative regulators of T cell activation. METHODS In view of the rapid and extensive development of this research field, we conducted a comprehensive review of the literature and update on the use of CTLA-4, PD-1 and PD-L1 targeted therapy in the treatment of several types of cancer including melanoma, non-small-cell lung carcinoma, breast cancer, hepatocellular carcinoma, hodgkin lymphoma, cervical cancer, head and neck squamous cell carcinoma. RESULTS Based on the last updated list released on March 2019, seven ICIs are approved by the FDA including ipilimumab, pembrolizumab, nivolumab, atezolizumab, avelumab, durvalumab, and cemiplimab. CONCLUSION This review also highlighted the most common adverse effects caused by ICIs and which affect people in different ways.
Collapse
Affiliation(s)
- Wissam Zam
- Department of Analytical and Food Chemistry, Faculty of Pharmacy, Al-Wadi International University, Homs. Syrian Arab Republic
| | - Lina Ali
- Department of Analytical and Food Chemistry, Faculty of Pharmacy, Tartous University, Tartous. Syrian Arab Republic
| |
Collapse
|
349
|
Should I Have Adjuvant Immunotherapy? An Interview Study Among Adults with Resected Stage 3 Melanoma and Their Partners. PATIENT-PATIENT CENTERED OUTCOMES RESEARCH 2021; 14:635-647. [PMID: 33759137 DOI: 10.1007/s40271-021-00507-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/28/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Adjuvant immunotherapy is a new treatment paradigm for adults with resected stage 3 melanoma. However, therapy can lead to long-term adverse health impacts, making immunotherapy decisions difficult. This study aimed to explore patients and their partners' views when considering whether to commence adjuvant immunotherapy. METHODS Focus groups and in-depth interviews were conducted among adults with resected stage 3 melanoma and their partners between August 2019 and April 2020. Factors important to adjuvant immunotherapy decision making were explored. Recruitment continued until data saturation, with thematic analysis performed. RESULTS Thirty-six participants were recruited across two cohorts, including 24 patients (mean age 65 years, 71% male), and 12 partners (mean age 69 years, 75% female). Twenty-two patients (92%) received adjuvant immunotherapy, two (8%) declined. Five patients (21%) ceased treatment early because of toxicity. Five themes about adjuvant immunotherapy were common to all participants: (1) life and death; (2) perceived risks and benefits; (3) seeking information; (4) healthcare team relationship; and (5) immunotherapy treatment considerations. Prolonging life was the primary consideration, with secondary concerns about treatment burden, timing, costs and efficacy. CONCLUSIONS This information can be used by clinicians to support melanoma treatment decision making.
Collapse
|
350
|
One-Year Morbidity Following Videoscopic Inguinal Lymphadenectomy for Stage III Melanoma. Cancers (Basel) 2021; 13:cancers13061450. [PMID: 33810068 PMCID: PMC8004993 DOI: 10.3390/cancers13061450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Inguinal lymphadenectomy (the removal of lymph nodes in the groin) is currently part of the treatment options for stage III melanoma patients. Surgery can be performed using one large inguinal incision (open approach) or a few smaller incisions (videoscopic approach). Previous research has already shown less severe complications and comparable oncologic outcomes after the videoscopic approach. Postoperative lymphedema following inguinal lymphadenectomy is a well-known problem which can potentially decrease quality of life. With the arrival of adjuvant systemic treatment options, less invalidating surgery is highly desirable. However, lymphedema and quality of life have only been investigated after the open approach. Therefore, we evaluated lymphedema and quality of life following videoscopic inguinal lymphadenectomy for stage III melanoma. The videoscopic inguinal lymphadenectomy is a feasible approach due to the comparable lymphedema incidence and normalization of quality of life during follow-up. Abstract Purpose: We aimed to elucidate morbidity following videoscopic inguinal lymphadenectomy for stage III melanoma. Methods: Melanoma patients who underwent a videoscopic inguinal lymphadenectomy between November 2015 and May 2019 were included. The measured outcomes were lymphedema and quality of life. Patients were reviewed one day prior to surgery and postoperatively every 3 months for one year. Results: A total number of 34 patients were included for participation; 19 (55.9%) patients underwent a concomitant iliac lymphadenectomy. Lymphedema incidence was 40% at 3 months and 50% at 12 months after surgery. Mean interlimb volume difference increased steadily from 1.8% at baseline to 6.9% at 12 months (p = 0.041). Median Lymph-ICF-LL total score increased from 0.0 at baseline to 12.0 at 3 months, and declined to 8.5 at 12 months (p = 0.007). Twelve months after surgery, Lymph-ICF-LL scores were higher for females (p = 0.021) and patients that received adjuvant radiotherapy (p = 0.013). The Median Distress Thermometer and EORTC QLQ-C30 summary score recovered to baseline at 12 months postoperatively (p = 0.747 and p = 0.203, respectively). Conclusions: The onset of lymphedema is rapid and continues to increase up to one year after videoscopic inguinal lymphadenectomy. Quality of life recovers to the baseline value.
Collapse
|