2301
|
Wu B, Shi L. Frontline BRAF Testing-Guided Treatment for Advanced Melanoma in the Era of Immunotherapies: A Cost-Utility Analysis Based on Long-term Survival Data. JAMA Dermatol 2020; 156:1177-1184. [PMID: 32697281 PMCID: PMC7376469 DOI: 10.1001/jamadermatol.2020.2398] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023]
Abstract
Importance The effectiveness of immune checkpoint inhibitors (ICIs) and BRAF and MEK inhibitors has improved advanced melanoma recovery. However, it is unknown whether these novel therapies are cost-effective for newly diagnosed advanced melanoma with unknown BRAF status. Objective To compare the cost-utility of these novel agents and their combinations with or without BRAF gene testing guidance for treating newly diagnosed advanced melanoma with unknown BRAF status. Design and Setting A decision-analytic model was adopted to project the outcomes of 8 strategies containing different ICIs and BRAF and MEK inhibitors for newly diagnosed advanced melanoma with unknown BRAF pathogenic variant status. The key clinical data were derived from the CheckMate 067, KEYNOTE-006, COMBI-d, and COMBI-v trials, and the cost and health preference data were derived from the literature. Costs were estimated from the US payer perspective. Main Outcomes and Measures Costs, quality-adjusted life-years (QALYs), incremental cost-utility ratio (ICUR), and incremental net health benefits were calculated. Subgroup, 1-way, and probabilistic sensitivity analyses were performed. Results Of the 8 competing strategies, nivolumab plus ipilimumab without patient selection based on BRAF pathogenic variant testing yielded the most significant health outcome, and the nivolumab strategy was the cheapest option. The nivolumab, pembrolizumab, and nivolumab plus ipilimumab strategies formed the cost-effective frontier, which showed the ordered ICURs were $8593 (SD, $592 995)/QALY for pembrolizumab vs nivolumab and $125 593 (SD, $5 751 223)/QALY for nivolumab plus ipilimumab vs pembrolizumab. Other strategies, including the BRAF testing-guided strategies (BRAF pathogenic variant testing followed by corresponding regimens for BRAF wild and pathogenic variant tumors), were dominated or extended dominated. The most influential parameters were the treatment efficacy of these new regimens. Conclusions and Relevance For newly diagnosed advanced melanoma with unknown BRAF pathogenic variant status, nivolumab plus ipilimumab and pembrolizumab strategies are likely to be the most cost-effective options. BRAF and MEK inhibitors might be productively placed in a second-line setting after BRAF pathogenic variant is confirmed.
Collapse
Affiliation(s)
- Bin Wu
- Medical Decision and Economic Group, Ren Ji Hospital, Department of Pharmacy, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lizheng Shi
- Department of Global Health Management and Policy, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| |
Collapse
|
2302
|
Krishnan T, Menzies AM, Roberts-Thomson R. Applying adjuvant therapy for melanoma into clinical practice. Expert Rev Anticancer Ther 2020; 21:129-133. [PMID: 33094675 DOI: 10.1080/14737140.2021.1841641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Tharani Krishnan
- Medical Oncology department, The Queen Elizabeth Hospital, Adelaide, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia.,Medical Oncology Department, Royal North Shore and Mater Hospitals, Sydney, Australia
| | | |
Collapse
|
2303
|
Baylet A, Laclaverie M, Marchand L, Bordes S, Closs-Gonthier B, Delpy L. Immunotherapies in cutaneous pathologies: an overview. Drug Discov Today 2020; 26:248-255. [PMID: 33137480 DOI: 10.1016/j.drudis.2020.10.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 10/02/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
Skin is a vital protective organ, the main role of which is to provide a physical barrier and to prevent the entry of pathogens. Various pathologies, such as atopic dermatitis (AD), psoriasis (PSO), or skin cancers, can affect the skin, and all show a high and increasing prevalence. Many antibodies are currently used in the treatment of these diseases. However, various studies are underway for the development of new biologics directed against specific targets. In this review, we describe current biologics used in skin pathologies as well as antibodies in development. We also discuss various immunotherapy examples that use new delivery technologies, such as microneedle patch, nanoparticles (NPs), liposomes, or gel formulation.
Collapse
Affiliation(s)
- Audrey Baylet
- Unité Mixte de Recherche CNRS 7276 - INSERM U1262 - Université de Limoges, CBRS, 2 rue du Dr Marcland, 87025 Limoges, France; Silab R&D Department, Brive, France
| | | | | | | | | | - Laurent Delpy
- Unité Mixte de Recherche CNRS 7276 - INSERM U1262 - Université de Limoges, CBRS, 2 rue du Dr Marcland, 87025 Limoges, France.
| |
Collapse
|
2304
|
Poels K, van Leent MM, Boutros C, Tissot H, Roy S, Meerwaldt AE, Toner YC, Reiche ME, Kusters PJ, Malinova T, Huveneers S, Kaufman AE, Mani V, Fayad ZA, de Winther MP, Marabelle A, Mulder WJ, Robert C, Seijkens TT, Lutgens E. Immune Checkpoint Inhibitor Therapy Aggravates T Cell-Driven Plaque Inflammation in Atherosclerosis. JACC CardioOncol 2020; 2:599-610. [PMID: 34396271 PMCID: PMC8352210 DOI: 10.1016/j.jaccao.2020.08.007] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/20/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Immunotherapy has revolutionized cancer treatment. However, immune checkpoint inhibitors (ICIs) that target PD-1 (programmed cell death protein-1) and/or CTLA-4 (cytotoxic T lymphocyte-associated antigen-4) are commonly associated with acute immune-related adverse events. Accumulating evidence also suggests that ICIs aggravate existing inflammatory diseases. OBJECTIVES As inflammation drives atherosclerotic cardiovascular disease, we studied the propensity of short-term ICI therapy to aggravate atherosclerosis. METHODS We used 18F-FDG (2-deoxy-2-[fluorine-18]fluoro-D-glucose) positron emission tomography-computed tomography to detect macrophage-driven vascular and systemic inflammation in pembrolizumab and nivolumab/ipilimumab-treated melanoma patients. In parallel, atherosclerotic Ldlr -/- mice were treated with CTLA-4 and PD-1 inhibition to study the proinflammatory consequences of immune checkpoint inhibition. RESULTS ICI treatment did not affect 18F-FDG uptake in the large arteries, spleen, and bone marrow of melanoma patients, nor myeloid cell activation in blood and lymphoid organs in hyperlipidemic mice. In contrast, we found marked changes in the adaptive immune response (i.e., increased CD4+ effector T cell and CD8+ cytotoxic T cell numbers in lymphoid organs and the arterial wall of our hyperlipidemic mice). Although plaque size was unaffected, plaques had progressed toward a lymphoid-based inflammatory phenotype, characterized by a 2.7-fold increase of CD8+ T cells and a 3.9-fold increase in necrotic core size. Increased endothelial activation was observed with a 2.2-fold and 1.6-fold increase in vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, respectively. CONCLUSIONS This study demonstrates that combination therapy with anti-CTLA-4 and anti-PD-1 antibodies does not affect myeloid-driven vascular and systemic inflammation in melanoma patients and hyperlipidemic mice. However, short-term ICI therapy in mice induces T cell-mediated plaque inflammation and drives plaque progression.
Collapse
Affiliation(s)
- Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Mandy M.T. van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Celine Boutros
- Oncology Department, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Hubert Tissot
- Radiology Department, Institut de Cancérologie Gustave Roussy, Paris, France
| | - Séverine Roy
- Oncology Department, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Anu E. Meerwaldt
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Yohana C.A. Toner
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Myrthe E. Reiche
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Pascal J.H. Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Tsveta Malinova
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Audrey E. Kaufman
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Venkatesh Mani
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zahi A. Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Aurelien Marabelle
- Oncology Department, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Willem J.M. Mulder
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Caroline Robert
- Oncology Department, Institut de Cancérologie Gustave Roussy, Villejuif, France
- Faculty of Medicine, Universite Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Tom T.P. Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Institute for Cardiovascular Prevention, Ludwig Maximilian University of Munich, Munich, Germany
- German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
2305
|
Pai SI, Faquin WC, Sadow PM, Pittet MJ, Weissleder R. New technology on the horizon: Fast analytical screening technique FNA (FAST-FNA) enables rapid, multiplex biomarker analysis in head and neck cancers. Cancer Cytopathol 2020; 128:782-791. [PMID: 32841527 PMCID: PMC8276888 DOI: 10.1002/cncy.22305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 01/26/2023]
Abstract
PD-L1 profiling was recently approved by the US Food and Drug Administration as a companion diagnostic for anti-PD1 treatment in patients with head and neck cancer, ushering in a new era for precision medicine. However, the routine development and implementation of such testing is still limited by current clinical workflows and the lack of better and more comprehensive alternatives. In this review, the authors discuss the real-world challenges of clinically based biomarker testing and highlight the advantages of developing fine-needle aspiration (FNA)-based biomarker testing that would enable frequent and serial tumor sampling. A conceptual and technological innovation is introduced, fast analytical screening technique (FAST)-FNA (FAST chemistry-enabled FNA), which is being developed to inform immunotherapy treatment options in patients with head and neck cancer and to assist with the development of the next generation of predictive biomarkers.
Collapse
Affiliation(s)
- Sara I. Pai
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - William C. Faquin
- Division of Head and Neck Pathology, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter M. Sadow
- Division of Head and Neck Pathology, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mikael J. Pittet
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
2306
|
Bergholz JS, Wang Q, Kabraji S, Zhao JJ. Integrating Immunotherapy and Targeted Therapy in Cancer Treatment: Mechanistic Insights and Clinical Implications. Clin Cancer Res 2020; 26:5557-5566. [PMID: 32576627 PMCID: PMC7641965 DOI: 10.1158/1078-0432.ccr-19-2300] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/08/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022]
Abstract
Small-molecule targeted therapies have demonstrated outstanding potential in the clinic. These drugs are designed to minimize adverse effects by selectively attacking cancer cells while exerting minimal damage to normal cells. Although initial response to targeted therapies may be high, yielding positive response rates and often improving survival for an important percentage of patients, resistance often limits long-term effectiveness. On the other hand, immunotherapy has demonstrated durable results, yet for a limited number of patients. Growing evidence indicates that some targeted agents can modulate different components of the antitumor immune response. These include immune sensitization by inhibiting tumor cell-intrinsic immune evasion programs or enhancing antigenicity, as well as direct effects on immune effector and immunosuppressive cells. The combination of these two approaches, therefore, has the potential to result in synergistic and durable outcomes for patients. In this review, we focus on the latest advances on integrating immunotherapy with small-molecule targeted inhibitors. In particular, we discuss how specific oncogenic events differentially affect immune response, and the implications of these findings on the rational design of effective combinations of immunotherapy and targeted therapies.
Collapse
Affiliation(s)
- Johann S Bergholz
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Qiwei Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Sheheryar Kabraji
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
2307
|
Ghebriou D, Prebet C, Bonnet G, Benderra MA. [New therapies in oncogeriatrics]. SOINS. GÉRONTOLOGIE 2020; 26:16-19. [PMID: 33549236 DOI: 10.1016/j.sger.2020.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cancer management is changing rapidly. Changes in practices are not all transferable to the elderly population, which is heterogeneous. The description of the intrinsic toxicity of anti-cancer treatments is insufficient in the elderly. Recent studies dedicated to the elderly incorporate composite evaluation criteria combining efficacy and toxicity with a broad definition including, among other things, loss of functional autonomy. These new data acquired, as well as new organisations integrating the new profession of advanced practice nurse in oncogeriatrics will enable us to better respond to the challenge of caring for elderly patients in the future.
Collapse
Affiliation(s)
- Djamel Ghebriou
- Oncologie médicale, site Tenon, Institut universitaire de cancérologie, Assistance publique-Hôpitaux de Paris, Sorbonne université, unité de coordination et antennes d'oncogériatrie Île-de-France-Paris-Est, 20 rue de la Chine, 75020 Paris, France.
| | - Coralie Prebet
- Oncologie médicale, site Tenon, Institut universitaire de cancérologie, Assistance publique-Hôpitaux de Paris, Sorbonne université, unité de coordination et antennes d'oncogériatrie Île-de-France-Paris-Est, 20 rue de la Chine, 75020 Paris, France
| | - Guillaume Bonnet
- Unité de coordination en oncogériatrie de Picardie, centre hospitalier universitaire Amiens-Picardie, site Sud, entrée secondaire, 30 avenue de la Croix-Jourdain, 80054 Amiens cedex 1, France
| | - Marc Antoine Benderra
- Oncologie médicale, site Tenon, Institut universitaire de cancérologie, Assistance publique-Hôpitaux de Paris, Sorbonne université, unité de coordination et antennes d'oncogériatrie Île-de-France-Paris-Est, 20 rue de la Chine, 75020 Paris, France
| |
Collapse
|
2308
|
Yang C, Sanchez-Vega F, Chang JC, Chatila WK, Shoushtari AN, Ladanyi M, Travis WD, Busam KJ, Rekhtman N. Lung-only melanoma: UV mutational signature supports origin from occult cutaneous primaries and argues against the concept of primary pulmonary melanoma. Mod Pathol 2020; 33:2244-2255. [PMID: 32581366 PMCID: PMC8386291 DOI: 10.1038/s41379-020-0594-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 01/14/2023]
Abstract
Primary pulmonary melanoma (PPM) is an entity recognized by the thoracic WHO classification. However, given the absence of native melanocytes in the lung and the known phenomenon of regression of cutaneous melanomas, the existence of PPM has remained controversial. Herein we investigate clinicopathologic and genomic features of lung-only melanomas with the goal to clarify their site of origin. We identified 10 melanomas involving exclusively lung with no current or previous cutaneous, uveal, or mucosal primaries. Four patients had solitary lesions with mean size of 5.1 cm (range 3.0-10.1 cm), meeting the criteria of PPM. Four patients had 2-3 lesions and 2 patients had >10 lesions. All cases underwent targeted next-generation sequencing interrogating up to 468 cancer genes, which revealed mean tumor mutation burden of 42.6 per megabase (range 1.8 to 126) and frequent mutations involving BRAF, NRAS, NF1, KIT, and KRAS - a genomic profile typical of UV-associated cutaneous melanoma. Mutational signature was assessable for eight cases harboring >20 mutations. This revealed that all evaluable cases harbored a dominant UV signature. In addition, one nonevaluable case harbored a GG > AA TERT promoter variant that is highly specific for UV-mutagenesis. As control groups, using the same methodology, a dominant UV signature was identified in 97% (470/486) of cutaneous melanomas, whereas no lung adenocarcinoma (n = 291) exhibited this signature. Notably, the clinical and pathologic features of solitary melanomas, especially those with large size and epithelioid morphology, closely mimicked primary lung carcinomas, highlighting a major potential for misdiagnosis. In conclusion, presence of a UV signature provides direct evidence that nearly all lung-only melanomas in this series, including solitary lesions meeting the strict criteria of PPM, represent metastases from occult cutaneous melanomas. This suggests that lung-only melanomas should be considered as likely metastatic even in the absence of a known primary melanoma elsewhere.
Collapse
Affiliation(s)
- Chen Yang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Walid K Chatila
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Klaus J Busam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2309
|
Keilholz U, Ascierto PA, Dummer R, Robert C, Lorigan P, van Akkooi A, Arance A, Blank CU, Chiarion Sileni V, Donia M, Faries MB, Gaudy-Marqueste C, Gogas H, Grob JJ, Guckenberger M, Haanen J, Hayes AJ, Hoeller C, Lebbé C, Lugowska I, Mandalà M, Márquez-Rodas I, Nathan P, Neyns B, Olofsson Bagge R, Puig S, Rutkowski P, Schilling B, Sondak VK, Tawbi H, Testori A, Michielin O. ESMO consensus conference recommendations on the management of metastatic melanoma: under the auspices of the ESMO Guidelines Committee. Ann Oncol 2020; 31:1435-1448. [PMID: 32763453 DOI: 10.1016/j.annonc.2020.07.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
Abstract
The European Society for Medical Oncology (ESMO) held a consensus conference on melanoma on 5-7 September 2019 in Amsterdam, The Netherlands. The conference included a multidisciplinary panel of 32 leading experts in the management of melanoma. The aim of the conference was to develop recommendations on topics that are not covered in detail in the current ESMO Clinical Practice Guideline and where available evidence is either limited or conflicting. The main topics identified for discussion were (i) the management of locoregional disease; (ii) targeted versus immunotherapies in the adjuvant setting; (iii) targeted versus immunotherapies for the first-line treatment of metastatic melanoma; (iv) when to stop immunotherapy or targeted therapy in the metastatic setting; and (v) systemic versus local treatment for brain metastases. The expert panel was divided into five working groups to each address questions relating to one of the five topics outlined above. Relevant scientific literature was reviewed in advance. Recommendations were developed by the working groups and then presented to the entire panel for further discussion and amendment before voting. This manuscript presents the results relating to the management of metastatic melanoma, including findings from the expert panel discussions, consensus recommendations and a summary of evidence supporting each recommendation. All participants approved the final manuscript.
Collapse
Affiliation(s)
- U Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - P A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - R Dummer
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - C Robert
- Department of Dermatology, Gustave Roussy, Villejuif, France; Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - P Lorigan
- Division of Cancer Sciences, The University of Manchester and The Christie NHS Foundation Trust, Manchester, UK
| | - A van Akkooi
- Department of Surgical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - A Arance
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - C U Blank
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - V Chiarion Sileni
- Department of Experimental and Clinical Oncology, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy
| | - M Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark; University of Copenhagen, Copenhagen, Denmark
| | - M B Faries
- Department of Surgery, The Angeles Clinic, Cedars Sinai Medical Center, Los Angeles, USA
| | - C Gaudy-Marqueste
- Department of Dermatology and Skin Cancer, Aix Marseille University, Hôpital De La Timone, Marseille, France
| | - H Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - J J Grob
- Department of Dermatology and Skin Cancer, Aix Marseille University, Hôpital De La Timone, Marseille, France
| | - M Guckenberger
- Department of Radio-Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - J Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A J Hayes
- Department of Academic Surgery, Royal Marsden NHS Foundation Trust, London, UK
| | - C Hoeller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - C Lebbé
- AP-HP Dermatology, Université de Paris, Paris, France; INSERM U976, Hôpital Saint Louis, Paris, France
| | - I Lugowska
- Early Phase Clinical Trials Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - M Mandalà
- Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | - I Márquez-Rodas
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - P Nathan
- Department of Medical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - B Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - R Olofsson Bagge
- Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Region Västra Götaland, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden
| | - S Puig
- Dermatology Service, Hospital Clínic of Barcelona and University of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; CIBER, Instituto de Salud Carlos III, Barcelona, Spain
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - B Schilling
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - V K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, USA
| | - H Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Testori
- Department of Dermatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - O Michielin
- Department of Oncology, University Hospital Lausanne, Lausanne, Switzerland
| |
Collapse
|
2310
|
Barrios DM, Do MH, Phillips GS, Postow MA, Akaike T, Nghiem P, Lacouture ME. Immune checkpoint inhibitors to treat cutaneous malignancies. J Am Acad Dermatol 2020; 83:1239-1253. [PMID: 32461079 PMCID: PMC7572574 DOI: 10.1016/j.jaad.2020.03.131] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
As the incidence of cutaneous malignancies continues to rise and their treatment with immunotherapy expands, dermatologists and their patients are more likely to encounter immune checkpoint inhibitors. While the blockade of immune checkpoint target proteins (cytotoxic T-lymphocyte-associated protein-4, programmed cell death-1, and programmed cell death ligand-1) generates an antitumor response in a substantial fraction of patients, there is a critical need for reliable predictive biomarkers and approaches to address refractory disease. The first article of this Continuing Medical Education series reviews the indications, efficacy, safety profile, and evidence supporting checkpoint inhibition as therapeutics for metastatic melanoma, cutaneous squamous cell carcinoma, and Merkel cell carcinoma. Pivotal studies resulting in the approval of ipilimumab, pembrolizumab, nivolumab, cemiplimab, and avelumab by regulatory agencies for various cutaneous malignancies, as well as ongoing clinical research trials, are discussed.
Collapse
Affiliation(s)
- Dulce M Barrios
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mytrang H Do
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medicine, New York, New York
| | - Gregory S Phillips
- State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Michael A Postow
- Weill Cornell Medicine, New York, New York; Melanoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tomoko Akaike
- Division of Dermatology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Paul Nghiem
- Division of Dermatology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Mario E Lacouture
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medicine, New York, New York.
| |
Collapse
|
2311
|
Cho SY, Huff DT, Jeraj R, Albertini MR. FDG PET/CT for Assessment of Immune Therapy: Opportunities and Understanding Pitfalls. Semin Nucl Med 2020; 50:518-531. [PMID: 33059821 PMCID: PMC8201415 DOI: 10.1053/j.semnuclmed.2020.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune checkpoint blockade has demonstrated the ability to modulate the immune system to produce durable responses in a wide range of cancers and has significantly impacted the standard of care. However, many cancer patients still do not respond to immune checkpoint blockade or have a limited duration of antitumor responses. Moreover, immune-related adverse events caused by immune checkpoint blockade can be severe and debilitating for some patients, limiting continuation of therapy and resulting in severe autoimmune conditions. Standard-of-care conventional anatomic imaging modalities and tumor response criteria have limitations to adequately assess tumor responses, especially early in the course of therapy, for risk-adapted clinical management to inform care of patients treated with immunotherapy. Molecular imaging with position emission tomography (PET) provides a noninvasive functional biomarker of tumor response, and of immune activation, for patients on immune-based therapies to help address these needs. 18F-FDG (FDG) PET/CT is readily available clinically and a number of studies have evaluated the use of this agent for assessment of prognosis, treatment response and immune activation for patients treated with immune checkpoint blockade. In this review paper, we discuss the current oncologic applications and imaging needs of cancer immunotherapy, recent studies applying FDG PET/CT for tumor response assessment, and evaluation of immune-related adverse events for improving clinical management. We largely focus on metastatic melanoma; however, we generalize where applicable to immunotherapy in other tumor types. We also briefly discuss PET imaging and quantitation as well as emerging non-FDG PET imaging radiotracers for cancer immunotherapy imaging.
Collapse
Affiliation(s)
- Steve Y Cho
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| | - Daniel T Huff
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Robert Jeraj
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI; Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
2312
|
Namikawa K, Kiyohara Y, Takenouchi T, Uhara H, Uchi H, Yoshikawa S, Takatsuka S, Koga H, Wada N, Minami H, Hatsumichi M, Namba Y, Yamazaki N. Final analysis of a phase II study of nivolumab in combination with ipilimumab for unresectable chemotherapy-naive advanced melanoma. J Dermatol 2020; 47:1257-1266. [PMID: 32812243 PMCID: PMC7693067 DOI: 10.1111/1346-8138.15514] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023]
Abstract
Nivolumab plus ipilimumab combination is currently one of the preferred regimens for advanced melanoma in recently updated clinical practice guidelines. However, the evidence on the efficacy of the combination for acral or mucosal subtypes remains less robust. This is the final analysis of a multicenter, open-label, uncontrolled phase II study that investigated the long-term efficacy and safety in treatment-naive Japanese patients with advanced melanoma, including acral or mucosal subtypes, and subsequent therapy after discontinuation of the investigational agents. Patients received four doses of nivolumab (1 mg/kg i.v.) in combination with ipilimumab (3 mg/kg i.v.) at 3-week intervals, followed by doses of nivolumab (3 mg/kg i.v.) at 2-week intervals. The median follow-up period was 20.8 months (range, 5.2-35.0). The centrally and locally assessed objective response rates were both 43.3% (13/30; 95% confidence interval [CI], 25.5-62.6). Median progression-free survival was not reached (95% CI, 3.02-not reached), and median overall survival was also not reached (95% CI, 19.52-not reached). The 30-month progression-free survival and overall survival rates were 50.3% and 54.2%, respectively. No new safety concerns were detected. After discontinuation of the investigational agents, 83.3% of patients received some form of subsequent therapy including 43.3% of patients who received nivolumab monotherapy and 26.7% of patients who received radiotherapy. Of the four patients who discontinued the investigational agents because of immune-related adverse events, two received subsequent therapy (nivolumab and ipilimumab, respectively) and the other two showed long-term treatment-free survival (659 and 590 days, respectively). Long-term survival with nivolumab plus ipilimumab was observed in Japanese patients with melanoma including acral and mucosal subtypes, which is consistent with the CheckMate 067 study. Many patients continued to receive some form of treatment safely after stopping treatment with nivolumab plus ipilimumab.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Yoshio Kiyohara
- Dermatology DivisionShizuoka Cancer Center HospitalShizuokaJapan
| | | | - Hisashi Uhara
- Department of DermatologyShinshu University School of MedicineMatsumotoJapan
- Present address:
Department of DermatologySapporo Medical UniversitySapporoJapan
| | - Hiroshi Uchi
- Department of DermatologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | | | - Sumiko Takatsuka
- Department of DermatologyNiigata Cancer Center HospitalNiigataJapan
| | - Hiroshi Koga
- Department of DermatologyShinshu University School of MedicineMatsumotoJapan
| | - Naoko Wada
- Department of DermatologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Hironobu Minami
- Department of Medical Oncology/HematologyKobe University Graduate School of MedicineKobeJapan
| | | | | | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
2313
|
Le Louedec F, Leenhardt F, Marin C, Chatelut É, Evrard A, Ciccolini J. Cancer Immunotherapy Dosing: A Pharmacokinetic/Pharmacodynamic Perspective. Vaccines (Basel) 2020; 8:E632. [PMID: 33142728 PMCID: PMC7712135 DOI: 10.3390/vaccines8040632] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Immune check-point inhibitors are drugs that are markedly different from other anticancer drugs because of their indirect mechanisms of antitumoral action and their apparently random effect in terms of efficacy and toxicity. This marked pharmacodynamics variability in patients calls for reconsidering to what extent approved dosing used in clinical practice are optimal or whether they should require efforts for customization in outlier patients. To better understand whether or not dosing could be an actionable item in oncology, in this review, preclinical and clinical development of immune checkpoint inhibitors are described, particularly from the angle of dose finding studies. Other issues in connection with dosing issues are developed, such as the flat dosing alternative, the putative role therapeutic drug monitoring could play, the rise of combinatorial strategies, and pharmaco-economic aspects.
Collapse
Affiliation(s)
- Félicien Le Louedec
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, and Cancer Research Center of Toulouse (CRCT), Inserm U1037, University of Toulouse, 31100 Toulouse, France;
| | - Fanny Leenhardt
- Institut de Cancérologie de Montpellier (ICM) and Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, University of Montpellier, 34090 Montpellier, France;
| | - Clémence Marin
- Assistance Publique—Hôpitaux de Marseille (AP-HM) and Simulation Modeling Adaptive Response for Therapeutics in cancer (SMARTc), Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France; (C.M.); (J.C.)
| | - Étienne Chatelut
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, and Cancer Research Center of Toulouse (CRCT), Inserm U1037, University of Toulouse, 31100 Toulouse, France;
| | - Alexandre Evrard
- Centre Hospitalier Universitaire de Nîmes Carémeau, Nîmes, France and IRCM U1194, University of Montpellier, 34090 Montpellier, France;
| | - Joseph Ciccolini
- Assistance Publique—Hôpitaux de Marseille (AP-HM) and Simulation Modeling Adaptive Response for Therapeutics in cancer (SMARTc), Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France; (C.M.); (J.C.)
| |
Collapse
|
2314
|
Dragomir MP, Moisoiu V, Manaila R, Pardini B, Knutsen E, Anfossi S, Amit M, Calin GA. A Holistic Perspective: Exosomes Shuttle between Nerves and Immune Cells in the Tumor Microenvironment. J Clin Med 2020; 9:jcm9113529. [PMID: 33142779 PMCID: PMC7693842 DOI: 10.3390/jcm9113529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
One of the limitations of cancer research has been the restricted focus on tumor cells and the omission of other non-malignant cells that are constitutive elements of this systemic disease. Current research is focused on the bidirectional communication between tumor cells and other components of the tumor microenvironment (TME), such as immune and endothelial cells, and nerves. A major success of this bidirectional approach has been the development of immunotherapy. Recently, a more complex landscape involving a multi-lateral communication between the non-malignant components of the TME started to emerge. A prime example is the interplay between immune and endothelial cells, which led to the approval of anti-vascular endothelial growth factor-therapy combined with immune checkpoint inhibitors and classical chemotherapy in non-small cell lung cancer. Hence, a paradigm shift approach is to characterize the crosstalk between different non-malignant components of the TME and understand their role in tumorigenesis. In this perspective, we discuss the interplay between nerves and immune cells within the TME. In particular, we focus on exosomes and microRNAs as a systemic, rapid and dynamic communication channel between tumor cells, nerves and immune cells contributing to cancer progression. Finally, we discuss how combinatorial therapies blocking this tumorigenic cross-talk could lead to improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Mihnea P. Dragomir
- Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Institute of Pathology, Charité University Hospital, 10117 Berlin, Germany
- Correspondence: (M.P.D.); (G.A.C.)
| | - Vlad Moisoiu
- Faculty of Physics, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania;
| | - Roxana Manaila
- Clinical Institute of Urology and Renal Transplantation, 400006 Cluj-Napoca, Romania;
| | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), 10060 Candiolo, Italy;
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT—The Arctic University of Norway, N-9037 Tromsø, Norway;
| | - Simone Anfossi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (M.P.D.); (G.A.C.)
| |
Collapse
|
2315
|
Vanneste BG, Van Limbergen EJ, Dubois L, Samarska IV, Wieten L, Aarts MJ, Marcelissen T, De Ruysscher D. Immunotherapy as sensitizer for local radiotherapy. Oncoimmunology 2020; 9:1832760. [PMID: 33194319 PMCID: PMC7605354 DOI: 10.1080/2162402x.2020.1832760] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/28/2022] Open
Abstract
The purpose of this report was to systematically review the radiation enhancement factor (REF) effects of immunotherapy on radiotherapy (RT) to the local tumor in comparison with other traditional radiation sensitizers such as cisplatin. PubMed and Medline databases were searched until February 2019. Reports with abscopal effect in the results were excluded. Graphs of the selected papers were digitized using Plot Digitizer (Sourceforge.net) in order to calculate the tumor growth delay (TGD) caused by immunotherapy. To enable comparison between different studies,the TGD were used to define the REF between RT versus the RT/immunotherapy combination. Thirty-two preclinical papers, and nine clinical series were selected. Different mouse models were exposed to RT doses ranging from 1 to 10 fractions of 1.8 to 20 Gray (Gy) per fraction. Endpoints were heterogeneous, ranging from regression to complete local response. No randomized clinical studies were identified. The median preclinical REF effect of different immunotherapy was varying from 1.7 to 9.1. There was no relationship observed either with subclasses of immunotherapy orRT doses. In the clinical studies, RT doses ranged from 1 to 37 fractions of 1.8 to 24 Gy per fraction. Most clinical trials used ipilimumab and interleukin-2. Local control rate in the clinical series ranged from 66% to 100%. A strong REF of immunotherapy (1.7 to 9.1) was observed, this being higher than traditionally sensitizers such as cisplatin (1.1). This result implies that for the same RT dose, a higher local control was achieved with a combination of immunotherapy and RT in preclinical settings. This study therefore supports the use of combined RT and immunotherapy to improve local tumor control in clinical settings without exacerbation of toxicities.
Collapse
Affiliation(s)
- Ben G.L. Vanneste
- Department of Radiation Oncology (MAASTRO Clinic), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Evert J Van Limbergen
- Department of Radiation Oncology (MAASTRO Clinic), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ludwig Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Iryna V. Samarska
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - L. Wieten
- Department of Transplantation Immunology, Tissue Typing Laboratory, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - M. J.B. Aarts
- Department of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - T. Marcelissen
- Department of Urology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO Clinic), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
2316
|
Pelster MS, Gruschkus SK, Bassett R, Gombos DS, Shephard M, Posada L, Glover MS, Simien R, Diab A, Hwu P, Carter BW, Patel SP. Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: Results From a Single-Arm Phase II Study. J Clin Oncol 2020; 39:599-607. [PMID: 33125309 DOI: 10.1200/jco.20.00605] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Metastatic uveal melanoma has poor overall survival (OS) and no approved systemic therapy options. Studies of single-agent immunotherapy regimens have shown minimal benefit. There is the potential for improved responses with the use of combination immunotherapy. PATIENTS AND METHODS We conducted a phase II study of nivolumab with ipilimumab in patients with metastatic uveal melanoma. Any number of prior treatments was permitted. Patients received nivolumab 1 mg/kg and ipilimumab 3 mg/kg for four cycles, followed by nivolumab maintenance therapy for up to 2 years. The primary outcome of the study was overall response rate (ORR) as determined by RECIST 1.1 criteria. Progression-free survival (PFS), OS, and adverse events were also assessed. RESULTS Thirty-five patients were enrolled, and 33 patients were evaluable for efficacy. The ORR was 18%, including one confirmed complete response and five confirmed partial responses. The median PFS was 5.5 months (95% CI, 3.4 to 9.5 months), and the median OS was 19.1 months (95% CI, 9.6 months to NR). Forty percent of patients experienced a grade 3-4 treatment-related adverse event. CONCLUSION The combination regimen of nivolumab plus ipilimumab demonstrates activity in metastatic uveal melanoma, with deep and sustained confirmed responses.
Collapse
Affiliation(s)
- Meredith S Pelster
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen K Gruschkus
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dan S Gombos
- Department of Head and Neck Surgery, Section of Ophthalmology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael Shephard
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Liberty Posada
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Maura S Glover
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rinata Simien
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brett W Carter
- Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
2317
|
Gandhi S, Pandey MR, Attwood K, Ji W, Witkiewicz AK, Knudsen ES, Allen C, Tario JD, Wallace PK, Cedeno CD, Levis M, Stack S, Funchain P, Drabick JJ, Bucsek MJ, Puzanov I, Mohammadpour H, Repasky EA, Ernstoff MS. Phase I Clinical Trial of Combination Propranolol and Pembrolizumab in Locally Advanced and Metastatic Melanoma: Safety, Tolerability, and Preliminary Evidence of Antitumor Activity. Clin Cancer Res 2020; 27:87-95. [PMID: 33127652 DOI: 10.1158/1078-0432.ccr-20-2381] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/09/2020] [Accepted: 10/21/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Increased β-adrenergic receptor (β-AR) signaling has been shown to promote the creation of an immunosuppressive tumor microenvironment (TME). Preclinical studies have shown that abrogation of this signaling pathway, particularly β2-AR, provides a more favorable TME that enhances the activity of anti-PD-1 checkpoint inhibitors. We hypothesize that blocking stress-related immunosuppressive pathways would improve tumor response to immune checkpoint inhibitors in patients. Here, we report the results of dose escalation of a nonselective β-blocker (propranolol) with pembrolizumab in patients with metastatic melanoma. PATIENTS AND METHODS A 3 + 3 dose escalation study for propranolol twice a day with pembrolizumab (200 mg every 3 weeks) was completed. The primary objective was to determine the recommended phase II dose (RP2D). Additional objectives included safety, antitumor activity, and biomarker analyses. Responders were defined as patients with complete or partial response per immune-modified RECIST at 6 months. RESULTS Nine patients with metastatic melanoma received increasing doses of propranolol in cohorts of 10, 20, and 30 mg twice a day. No dose-limiting toxicities were observed. Most common treatment-related adverse events (TRAEs) were rash, fatigue, and vitiligo, observed in 44% patients. One patient developed two grade ≥3 TRAEs. Objective response rate was 78%. While no significant changes in treatment-associated biomarkers were observed, an increase in IFNγ and a decrease in IL6 was noted in responders. CONCLUSIONS Combination of propranolol with pembrolizumab in treatment-naïve metastatic melanoma is safe and shows very promising activity. Propranolol 30 mg twice a day was selected as RP2D in addition to pembrolizumab based on safety, tolerability, and preliminary antitumor activity.
Collapse
Affiliation(s)
- Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| | - Manu R Pandey
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Wenyan Ji
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Agnieszka K Witkiewicz
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Erik S Knudsen
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Cheryl Allen
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Joseph D Tario
- Department of Flow Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Paul K Wallace
- Department of Flow Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Carlos D Cedeno
- Department of Flow Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Maria Levis
- Clinical Research Service, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Suzanne Stack
- Clinical Research Service, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Pauline Funchain
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Joseph J Drabick
- Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania
| | - Mark J Bucsek
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Marc S Ernstoff
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York.,Division of Cancer Treatment and Diagnosis/Developmental Therapy Program-ImmunoOncology Branch, NIH/NCI, Bethesda, Maryland
| |
Collapse
|
2318
|
Liu H, Xu D, Wang W, Sun F, Zhang S, Yang X, Tian Y. Systematic Assessment of Risk of Fever in Solid Tumor Patients Treated With PD-1/PD-L1 Inhibitors: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:570080. [PMID: 33194659 PMCID: PMC7658543 DOI: 10.3389/fonc.2020.570080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Purpose: Drug-induced fever is frequently reported in cancer patients treated with anti-programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1), and stoppage of the offending agent is the management of choice. However, given the complex management of cancer patients, this needs to be carefully studied. Therefore, we conducted a meta-analysis to estimate the risk of fever associated with anti-PD-1/PD-L1 in cancer patients. Methods: From May 2010 to 2020, an electronic search was conducted through PubMed for relevant studies. All clinical trials reporting fever in cancer patients treated with PD-1/PD-L1 inhibitors were included, while other designs were excluded. A manual search was also conducted to search for relevant articles. Outcomes included the risk of pyrexia and febrile neutropenia in the overall population and based on the grade of fever (all grades vs. grades 3–5). The Newcastle–Ottawa Scale was used to assess the quality of included studies. Results: Thirty-one articles, involving 27 clinical trials and 15,867 participants, were included. The increased risk of pyrexia for all grades is only found when PD-1/PD-L1 plus cytotoxic T lymphocyte-associated protein 4 (CTLA-4) was compared to CTLA-4 [odds ratio (OR) = 2.48, 95% CI: 1.17, 5.23]. The risk of febrile neutropenia for all-grade fever was significantly lower in the PD-1/PD-L1 group compared to that of chemotherapy alone (OR = 0.02, 95% CI: 0.01, 0.05). A similar trend in the risk of febrile neutropenia was also found for grades 3–5 (OR = 0.02, 95% CI: 0.01, 0.05). Conclusion: The increased risk of pyrexia for all grades could only be found when PD-1/PD-L1 plus CTLA-4 was compared with CTLA-4. Meanwhile, compared to chemotherapy, PD-1/PD-L1 inhibitors reduced the risk of febrile neutropenia.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Radiotherapy Oncology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China
| | - Dongmei Xu
- Nursing Department, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China
| | - Wentao Wang
- Department of Liver Transplantation and Hepatic Surgery, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China
| | - Fengchao Sun
- Department of Pediatrics, The People's Hospital of Yuncheng County, Heze, China
| | - Shuisheng Zhang
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Xiaowei Yang
- Department of Hepatobiliary Intervention, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yuan Tian
- Department of Radiotherapy Oncology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China
| |
Collapse
|
2319
|
Lancelot M, Miller MJ, Roback J, Stowell SR. Refractory thrombotic thrombocytopenic purpura related to checkpoint inhibitor immunotherapy. Transfusion 2020; 61:322-328. [PMID: 33119913 DOI: 10.1111/trf.16117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Checkpoint inhibitors enhance T-lymphocyte-mediated antitumor responses, resulting in increased survival for patients with neoplastic disease. However, a subset of patients receiving checkpoint inhibitor therapy may experience adverse complications that include the development of autoimmune conditions, such as thrombotic thrombocytopenic purpura (TTP). Given the potential etiologic differences of checkpoint inhibitor-related autoimmunity, TTP that develops in the presence of checkpoint inhibitors may be refractory to current treatment methods and therefore may require additional treatment and prognostic consideration. CASE REPORT Herein, we describe the unique clinical course of a patient who was treated with the combined checkpoint inhibitors nivolumab and ipilimumab for Stage IV malignant melanoma, who subsequently developed TTP. Unlike many patients with TTP, this patient failed to develop a sustained response to therapeutic plasma exchange. Additional use of steroids, anti-CD20, and plasma cell-targeting therapy (bortezomib) also failed to substantially reverse thrombocytopenia in a sustainable fashion. During this time, her melanoma progressed, and she ultimately succumbed. CONCLUSION This case illustrates not only that TTP may be a potential complication of checkpoint inhibitor therapy, but also that TTP developing in this setting may result in an unpredictable response to commonly employed TTP treatment modalities. Ultimately, checkpoint inhibitor-related TTP may require distinct management approaches and prognostic considerations.
Collapse
Affiliation(s)
- Moira Lancelot
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maureen J Miller
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - John Roback
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sean R Stowell
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
2320
|
Wu M, Hu W, Wang G, Yao Y, Yu XF. Nicotinamide N-Methyltransferase Is a Prognostic Biomarker and Correlated With Immune Infiltrates in Gastric Cancer. Front Genet 2020; 11:580299. [PMID: 33193702 PMCID: PMC7655872 DOI: 10.3389/fgene.2020.580299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the third most common cause of cancer-related death in the word. Immunotherapy is a promising treatment of cancer. However, it is unclear which GC subpopulation would benefit most from immunotherapy and it is necessary to develop effective biomarkers for predicting immunotherapy response. Nicotinamide N-methyltransferase (NNMT) is a metabolic regulator of cancer-associated fibroblast (CAF) differentiation and cancer progression. In this study, we explored the correlations of NNMT to tumor-infiltrating immune cells (TIICs) and immune marker sets in The Cancer Genome Atlas Stomach Adenocarcinoma STAD (TCGA-STAD). Subsequently, we screened the NNMT correlated genes and performed the enrichment analysis of these genes. We eventually predicted the 19 most potential small-molecule drugs using the connectivity map (CMap) and Comparative Toxicogenomics Database (CTD). Also, nadolol, tranexamic acid, felbinac and dapsone were considered the four most promising drugs for GC. In summary, NNMT can be used as a prognostic biomarker that reflect immune infiltration level and a novel therapeutic target in GC.
Collapse
Affiliation(s)
- Miaowei Wu
- Cancer Institute, Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine, Hangzhou, China
| | - Weilei Hu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Guosheng Wang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yihan Yao
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Fang Yu
- Cancer Institute, Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2321
|
Abstract
Immunotherapy with immune checkpoint inhibition has dramatically changed the treatment of melanoma. Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 are approved for the treatment of advanced melanoma alone and in combination. In addition, these agents are approved for use in high-risk resected stage III melanoma in the adjuvant setting. Clinical trials testing the combination of immune checkpoint inhibition with other therapies and novel immunotherapies continue. This article reviews the current literature on approved uses of immune checkpoint inhibition in melanoma and discusses ongoing trials and future directions.
Collapse
|
2322
|
Abstract
End-of-life care of critically ill adult patients with advanced or incurable cancers is imbued with major ethical challenges. Oncologists, hospitalists, and intensivists can inadvertently subjugate themselves to the perceived powers of autonomous patients. Therapeutic illusion and poor insight by surrogates in physicians' ability to offer accurate prognosis, missed opportunities and miscommunication by clinicians, and lack of systematic or protocolized approach represent important barriers to high-quality palliative care. Enhanced collaboration, models that allow clinicians and surrogates to share the burdens of decision, and institutional support for early integration of palliative care can foster an ethical climate.
Collapse
Affiliation(s)
- Jamie C Riches
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Louis P Voigt
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
2323
|
van Sluis J, de Heer EC, Boellaard M, Jalving M, Brouwers AH, Boellaard R. Clinically feasible semi-automatic workflows for measuring metabolically active tumour volume in metastatic melanoma. Eur J Nucl Med Mol Imaging 2020; 48:1498-1510. [PMID: 33099667 PMCID: PMC8113298 DOI: 10.1007/s00259-020-05068-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/12/2020] [Indexed: 12/19/2022]
Abstract
Purpose Metabolically active tumour volume (MATV) is a potential quantitative positron emission tomography (PET) imaging biomarker in melanoma. Accumulating data indicate that low MATV may predict increased chance of response to immunotherapy and overall survival. However, metastatic melanoma can present with numerous (small) tumour lesions, making manual tumour segmentation time-consuming. The aim of this study was to evaluate multiple semi-automatic segmentation workflows to determine reliability and reproducibility of MATV measurements in patients with metastatic melanoma. Methods An existing cohort of 64 adult patients with histologically proven metastatic melanoma was used in this study. 18F-FDG PET/CT diagnostic baseline images were acquired using a European Association of Nuclear Medicine (EANM) Research Limited–accredited Siemens Biograph mCT PET/CT system (Siemens Healthineers, Knoxville, USA). PET data were analysed using manual, gradient-based segmentation and five different semi-automatic methods: three direct PET image–derived delineations (41MAX, A50P and SUV40) and two based on a majority-vote approach (MV2 and MV3), without and with (suffix ‘+’) manual lesion addition. Correlation between the different segmentation methods and their respective associations with overall survival was assessed. Results Correlation between the MATVs derived by the manual segmentation and semi-automated tumour segmentations ranged from R2 = 0.41 for A50P to R2 = 0.85 for SUV40+ and MV2+, respectively. Manual MATV segmentation did not differ significantly from the semi-automatic methods SUV40 (∆MATV mean ± SD 0.08 ± 0.60 mL, P = 0.303), SUV40+ (∆MATV − 0.10 ± 0.51 mL, P = 0.126), MV2+ (∆MATV − 0.09 ± 0.62 mL, P = 0.252) and MV3+ (∆MATV − 0.03 ± 0.55 mL, P = 0.615). Log-rank tests showed statistically significant overall survival differences between above and below median MATV patients for all segmentation methods with areas under the ROC curves of 0.806 for manual segmentation and between 0.756 [41MAX] and 0.807 [MV3+] for semi-automatic segmentations. Conclusions Simple and fast semi-automated FDG PET segmentation workflows yield accurate and reproducible MATV measurements that correlate well with manual segmentation in metastatic melanoma. The most readily applicable and user-friendly SUV40 method allows feasible MATV measurement in prospective multicentre studies required for validation of this potential PET imaging biomarker for clinical use. Electronic supplementary material The online version of this article (10.1007/s00259-020-05068-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Ellen C de Heer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Mayke Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands. .,Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Cancer Center Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
2324
|
Bleicher J, Swords DS, Mali ME, McGuire L, Pahlkotter MK, Asare EA, Bowles TL, Hyngstrom JR. Recurrence patterns in patients with Stage II melanoma: The evolving role of routine imaging for surveillance. J Surg Oncol 2020; 122:1770-1777. [PMID: 33098702 DOI: 10.1002/jso.26214] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES The relatively recent availability of effective systemic therapies for metastatic melanoma necessitates reconsideration of current surveillance patterns. Evidence supporting surveillance guidelines for resected Stage II melanoma is lacking. Prior reports note routine imaging detects only 21% of recurrent disease. This study aims to define recurrence patterns for Stage II melanoma to inform future surveillance guidelines. METHODS This is a retrospective study of patients with Stage II melanoma. We analyzed risk factors for recurrence and methods of recurrence detection. We also assessed survival. Yearly hazards of recurrence were visualized. RESULTS With a median follow-up of 4.9 years, 158 per 580 patients (27.2%) recurred. Overall, most recurrences were patient-detected (60.7%) or imaging-detected (27.3%). Routine imaging was important in detecting recurrence in patients with distant recurrences (adjusted rate 43.1% vs. 9.4% for local/in-transit; p = .04) and with Stage IIC melanoma (42.5% vs. 18.5% for IIA; p = .01). Male patients also self-detected recurrent disease less than females (52.1% vs. 76.8%; p < .01). CONCLUSIONS Routine imaging surveillance played a larger role in detecting recurrent disease for select groups in this cohort than noted in prior studies. In an era of effective systemic therapy, routine imaging should be considered for detection of asymptomatic relapse for select, high-risk patient groups.
Collapse
Affiliation(s)
- Josh Bleicher
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Douglas S Swords
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Meghan E Mali
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Lauren McGuire
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Maranda K Pahlkotter
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Elliot A Asare
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Surgery, Intermountain Medical Center, Salt Lake City, Utah, USA
| | - Tawnya L Bowles
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Surgery, Intermountain Medical Center, Salt Lake City, Utah, USA
| | - John R Hyngstrom
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Surgery, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
2325
|
Cheung VTF, Brain O. Immunotherapy induced enterocolitis and gastritis - What to do and when? Best Pract Res Clin Gastroenterol 2020; 48-49:101703. [PMID: 33317787 DOI: 10.1016/j.bpg.2020.101703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 01/31/2023]
Abstract
Oncological treatment has been revolutionised by the advent of immune checkpoint inhibitors (ICPi), which block inhibitory immune pathways to enhance anti-tumour responses and improve survival. This mode of action is non-specific so can cause immune-related adverse events, of which diarrhoea and enterocolitis are amongst the most common. ICPi-enterocolitis frequently leads to cancer therapy interruption. ICPi-gastritis typically occurs at a later stage of ICPi therapy and can present more insidiously with nausea and vomiting. ICPi-enterocolitis and gastritis are treated with corticosteroids, with refractory cases typically requiring biologic therapy. This review will briefly consider the pathogenesis of ICPi-induced GI disease, before focussing on the practical management of these conditions. The anticipated global increase in ICPi use across cancer types highlights the importance of prospective research in order that we can understand the immuno-microbiology of ICPi-enterocolitis and gastritis. This will lead to predictive biomarkers and help to define optimal treatment regimens.
Collapse
Affiliation(s)
- Vincent Ting Fung Cheung
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | - Oliver Brain
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
2326
|
Hu W, Wang G, Wang Y, Riese MJ, You M. Uncoupling Therapeutic Efficacy from Immune-Related Adverse Events in Immune Checkpoint Blockade. iScience 2020; 23:101580. [PMID: 33083746 PMCID: PMC7554032 DOI: 10.1016/j.isci.2020.101580] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy with monoclonal antibodies targeting immune checkpoint molecules, including programmed death-1 (PD-1), PD ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen (CTLA)-4, has become prominent in the treatment of many types of cancer. However, a significant number of patients treated with immune checkpoint inhibitors (ICIs) develop immune-related adverse events (irAEs). irAEs can affect any organ system, and although most are clinically manageable, irAEs can result in mortality or long-term morbidity. Factors that can predict irAEs remain elusive. Understanding the etiology of ICI-induced irAEs and ways to limit these adverse events are needed. In this review, we provide basic science and clinical insights on the mechanisms responsible for ICI efficacy and ICI-induced irAEs. We further provide insights into approaches that may uncouple irAEs from the ability of ICIs to kill tumor cells.
Collapse
Affiliation(s)
- Weilei Hu
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Guosheng Wang
- Department of Biomedical Engineering, Binghamton University—SUNY, 4400 Vestal Pkwy E, Binghamton, NY 13902, USA
| | - Yian Wang
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Blood Research Institute, Versiti Inc, Milwaukee, WI 53226, USA
| | - Ming You
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
2327
|
Pavlick AC, Zhao R, Lee CH, Ritchings C, Rao S. First-line immunotherapy versus targeted therapy in patients with BRAF-mutant advanced melanoma: a real-world analysis. Future Oncol 2020; 17:689-699. [PMID: 33084375 DOI: 10.2217/fon-2020-0643] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To compare effectiveness of nivolumab + ipilimumab (NIVO + IPI) versus BRAF + MEK inhibitors (BRAFi + MEKi) in patients with BRAF-mutant advanced melanoma in the real-world setting. Materials & methods: This study used the Flatiron Health electronic medical record database. Results: After adjusting for differences in baseline characteristics, NIVO + IPI was associated with a 32% reduction in risk of death versus BRAFi + MEKi. At a mean follow-up of 15-16 months, 64% of NIVO + IPI patients and 43% of BRAFi + MEKi patients were alive; subsequent therapy was administered to 33 and 41% of patients, respectively. After first-line NIVO + IPI, 20% of patients died before subsequent therapy, whereas 32% died after first-line BRAFi + MEKi. Conclusion: In this real-world study, patients treated with first-line NIVO + IPI showed significant survival benefit versus those receiving first-line BRAFi + MEKi.
Collapse
Affiliation(s)
| | - Ruizhi Zhao
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | - Cho-Han Lee
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | | | - Sumati Rao
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| |
Collapse
|
2328
|
Hoyek S, Kourie HR, Labaki C, Antoun J. Immune checkpoint inhibitors in ocular melanomas: contrasting efficacy with cutaneous melanomas. Immunotherapy 2020; 12:1149-1152. [PMID: 33076742 DOI: 10.2217/imt-2020-0234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Sandra Hoyek
- Ophtalmology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Hampig Raphael Kourie
- Hematology-Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Chris Labaki
- Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Joelle Antoun
- Ophtalmology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| |
Collapse
|
2329
|
Quagliariello V, De Laurentiis M, Cocco S, Rea G, Bonelli A, Caronna A, Lombari MC, Conforti G, Berretta M, Botti G, Maurea N. NLRP3 as Putative Marker of Ipilimumab-Induced Cardiotoxicity in the Presence of Hyperglycemia in Estrogen-Responsive and Triple-Negative Breast Cancer Cells. Int J Mol Sci 2020; 21:E7802. [PMID: 33096896 PMCID: PMC7589802 DOI: 10.3390/ijms21207802] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/10/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Hyperglycemia, obesity and metabolic syndrome are negative prognostic factors in breast cancer patients. Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, achieving unprecedented efficacy in multiple malignancies. However, ICIs are associated with immune-related adverse events involving cardiotoxicity. We aimed to study if hyperglycemia could affect ipilimumab-induced anticancer efficacy and enhance its cardiotoxicity. Human cardiomyocytes and estrogen-responsive and triple-negative breast cancer cells (MCF-7 and MDA-MB-231 cell lines) were exposed to ipilimumab under high glucose (25 mM); low glucose (5.5 mM); high glucose and co-administration of SGLT-2 inhibitor (empagliflozin); shifting from high glucose to low glucose. Study of cell viability and the expression of new putative biomarkers of cardiotoxicity and resistance to ICIs (NLRP3, MyD88, cytokines) were quantified through ELISA (Cayman Chemical) methods. Hyperglycemia during treatment with ipilimumab increased cardiotoxicity and reduced mortality of breast cancer cells in a manner that is sensitive to NLRP3. Notably, treatment with ipilimumab and empagliflozin under high glucose or shifting from high glucose to low glucose reduced significantly the magnitude of the effects, increasing responsiveness to ipilimumab and reducing cardiotoxicity. To our knowledge, this is the first evidence that hyperglycemia exacerbates ipilimumab-induced cardiotoxicity and decreases its anticancer efficacy in MCF-7 and MDA-MB-231 cells. This study sets the stage for further tests on other breast cancer cell lines and primary cardiomyocytes and for preclinical trials in mice aimed to decrease glucose through nutritional interventions or administration of gliflozines during treatment with ipilimumab.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (M.C.L.); (G.C.)
| | - Michelino De Laurentiis
- Breast Unit, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (M.D.L.); (S.C.)
| | - Stefania Cocco
- Breast Unit, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (M.D.L.); (S.C.)
| | - Giuseppina Rea
- UOC Bersagli Molecolari del Microambiente, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131 Naples, Italy;
| | - Annamaria Bonelli
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (M.C.L.); (G.C.)
| | - Antonietta Caronna
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (M.C.L.); (G.C.)
| | - Maria Cristina Lombari
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (M.C.L.); (G.C.)
| | - Gabriele Conforti
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (M.C.L.); (G.C.)
| | - Massimiliano Berretta
- Department of MedicalOncology-Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (M.C.L.); (G.C.)
| |
Collapse
|
2330
|
Wu CE, Yang CK, Peng MT, Huang PW, Chang CF, Yeh KY, Chen CB, Wang CL, Hsu CW, Chen IW, Lin CT, Ueng SH, Lin G, Lin YF, Cheng CY, Chang JWC. The association between immune-related adverse events and survival outcomes in Asian patients with advanced melanoma receiving anti-PD-1 antibodies. BMC Cancer 2020; 20:1018. [PMID: 33087090 PMCID: PMC7579996 DOI: 10.1186/s12885-020-07508-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/07/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The association between immune-related adverse events (irAEs) and survival outcomes in patients with advanced melanoma receiving therapy with immune checkpoint inhibitors (ICIs) has not been well established, particularly in Asian melanoma. METHODS We retrospectively reviewed 49 melanoma patients undergoing therapy with ICIs (anti-PD-1 monotherapy), and analyzed the correlation between irAEs and clinical outcomes including progression-free survival (PFS) and overall survival (OS). RESULTS Overall, the patients who experienced grade 1-2 irAEs had longer PFS (median PFS, 4.6 vs. 2.5 months; HR, 0.52; 95% CI: 0.27-0.98; p = 0.042) and OS (median OS, 15.2 vs. 5.7 months; HR, 0.50; 95% CI: 0.24-1.02; p = 0.058) than the patients who did not experience irAEs. Regarding the type of irAE, the patients with either skin/vitiligo or endocrine irAEs showed better PFS (median PFS, 6.1 vs. 2.7 months; HR, 0.40, 95% CI: 0.21-0.74; p = 0.003) and OS (median OS, 18.7 vs. 4.5 months; HR, 0.34, 95% CI: 0.17-0.69, p = 0.003) than patients without any of these irAEs. CONCLUSIONS Melanoma patients undergoing anti-PD-1 monotherapy and experiencing mild-to-moderate irAEs (grade 1-2), particularly skin (vitiligo)/endocrine irAEs had favorable survival outcomes. Therefore, the association between irAEs and the clinical outcomes in melanoma patients undergoing anti-PD-1 ICIs may be severity and type dependent.
Collapse
Affiliation(s)
- Chiao-En Wu
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan, Taiwan.
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Chan-Keng Yang
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan, Taiwan
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Meng-Ting Peng
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan, Taiwan
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Pei-Wei Huang
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan, Taiwan
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ching-Fu Chang
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan, Taiwan
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Kun-Yun Yeh
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Division of Hemato-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chun-Bing Chen
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital at Linkou, Taipei, Keelung, Taiwan
| | - Chih-Liang Wang
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chao-Wei Hsu
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - I-Wen Chen
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Cheng-Tao Lin
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, , Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Shir-Hwa Ueng
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Gigin Lin
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Medical Imaging & Intervention, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yu-Fen Lin
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Nursing, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chi-Yuan Cheng
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Pharmacy, , Chang Gung Memorial Hospital, Linkou, Taiwan
| | - John Wen-Cheng Chang
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Chang Gung University College of Medicine, 5, Fu-Hsing Street, Kwei-Shan, Taoyuan, Taiwan.
- Immuno-Oncology Center of Excellence, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| |
Collapse
|
2331
|
Das I, Chen H, Maddalo G, Tuominen R, Rebecca VW, Herlyn M, Hansson J, Davies MA, Egyházi Brage S. Inhibiting insulin and mTOR signaling by afatinib and crizotinib combination fosters broad cytotoxic effects in cutaneous malignant melanoma. Cell Death Dis 2020; 11:882. [PMID: 33082316 PMCID: PMC7576205 DOI: 10.1038/s41419-020-03097-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/27/2022]
Abstract
Current treatment modalities for disseminated cutaneous malignant melanoma (CMM) improve survival, however disease progression commonly ensues. In a previous study we identified afatinib and crizotinib in combination as a novel potential therapy for CMM independent of BRAF/NRAS mutation status. Herein, we elucidate the underlying mechanisms of the combination treatment effect to find biomarkers and novel targets for development of therapy that may provide clinical benefit by proteomic analysis of CMM cell lines and xenografts using mass spectrometry based analysis and reverse phase protein array. Identified candidates were validated using immunoblotting or immunofluorescence. Our analysis revealed that mTOR/Insulin signaling pathways were significantly decreased by the afatinib and crizotinib combination treatment. Both in vitro and in vivo analyses showed that the combination treatment downregulated pRPS6KB1 and pRPS6, downstream of mTOR signaling, and IRS-1 in the insulin signaling pathway, specifically ablating IRS-1 nuclear signal. Silencing of RPS6 and IRS-1 alone had a similar effect on cell death, which was further induced when IRS-1 and RPS6 were concomitantly silenced in the CMM cell lines. Silencing of IRS-1 and RPS6 resulted in reduced sensitivity towards combination treatment. Additionally, we found that IRS-1 and RPS6KB1 expression levels were increased in advanced stages of CMM clinical samples. We could demonstrate that induced resistance towards combination treatment was reversible by a drug holiday. CD171/L1CAM, mTOR and PI3K-p85 were induced in the combination resistant cells whereas AXL and EPHA2, previously identified mediators of resistance to MAPK inhibitor therapy in CMM were downregulated. We also found that CD171/L1CAM and mTOR were increased at progression in tumor biopsies from two matched cases of patients receiving targeted therapy with BRAFi. Overall, these findings provide insights into the molecular mechanisms behind the afatinib and crizotinib combination treatment effect and leverages a platform for discovering novel biomarkers and therapy regimes for CMM treatment.
Collapse
Affiliation(s)
- Ishani Das
- Department of Oncology-Pathology, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - Huiqin Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gianluca Maddalo
- Science for Life Laboratory, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rainer Tuominen
- Department of Oncology-Pathology, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - Vito W Rebecca
- Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - Michael A Davies
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
2332
|
Quagliariello V, Passariello M, Rea D, Barbieri A, Iovine M, Bonelli A, Caronna A, Botti G, De Lorenzo C, Maurea N. Evidences of CTLA-4 and PD-1 Blocking Agents-Induced Cardiotoxicity in Cellular and Preclinical Models. J Pers Med 2020; 10:jpm10040179. [PMID: 33086484 PMCID: PMC7711520 DOI: 10.3390/jpm10040179] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Several strategies based on immune checkpoint inhibitors (ICIs) have been developed for cancer therapy, opening to advantages in cancer outcomes. However, several ICI-induced side effects have emerged in these patients, especially a rare but clinically significant cardiotoxicity with high rate of mortality. We studied the cytotoxic and pro-inflammatory properties of Ipilimumab and Nivolumab, the underlying pathways and cytokine storm involved. Methods: Co-cultures of human cardiomyocytes and lymphocytes were exposed to Ipilimumab or Nivolumab; cell viability and expression of leukotrienes, NLRP3, MyD88, and p65/NF-kB were performed. C57 mice were treated with Ipilimumab (15 mg/kg); analysis of fractional shortening, ejection fraction, radial and longitudinal strain were made before and after treatments through 2D-echocardiography. Expression of NLRP3, MyD88, p65/NF-kB, and 12 cytokines were analyzed in murine myocardium. Results: Nivolumab and Ipilimumab exert effective anticancer, but also significant cardiotoxic effects in co-cultures of lymphocytes and tumor or cardiac cells. Both ICIs increased NLRP3, MyD88, and p65/NF-kB expression compared to untreated cells, however, the most pro-inflammatory and cardiotoxic effects were seen after exposure to Ipilimumab. Mice treated with Ipilimumab showed a significant decrease in fractional shortening and radial strain with respect to untreated mice, coupled with a significant increase in myocardial expression of NLRP3, MyD88, and several interleukins. Conclusions: Nivolumab and Ipilimumab exert cytotoxic effects mediated by the NLRP3/IL-1β and MyD88 pathways, leading to pro-inflammatory cytokine storm in heart tissue.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | | | - Domenica Rea
- Animal Facility, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (D.R.); (A.B.)
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (D.R.); (A.B.)
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Annamaria Bonelli
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Antonietta Caronna
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Claudia De Lorenzo
- CEINGE—Biotecnologie Avanzate s.c.a.r.l., 80131 Naples, Italy;
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Napoli, Italy
- Correspondence: (C.D.L.); (N.M.)
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
- Correspondence: (C.D.L.); (N.M.)
| |
Collapse
|
2333
|
Aoude LG, Bonazzi VF, Brosda S, Patel K, Koufariotis LT, Oey H, Nones K, Wood S, Pearson JV, Lonie JM, Arneil M, Atkinson V, Smithers BM, Waddell N, Barbour AP. Pathogenic germline variants are associated with poor survival in stage III/IV melanoma patients. Sci Rep 2020; 10:17687. [PMID: 33077847 PMCID: PMC7572377 DOI: 10.1038/s41598-020-74956-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Patients with late stage resected cutaneous melanoma have poor overall survival (OS) and experience irreversible adverse events from systemic therapy. There is a clinical need to identify biomarkers to predict outcome. Performing germline/tumour whole-exome sequencing of 44 stage III/IV melanoma patients we identified pathogenic germline mutations in CDKN2A, CDK4, ATM, POLH, MRE11A, RECQL4 and XPC, affecting 7/44 patients. These mutations were associated with poor OS (p = 0.0082). We confirmed our findings in The Cancer Genome Atlas (TCGA) human skin cutaneous melanoma cohort where we identified pathogenic variants in 40/455 patients (p = 0.0203). Combining these cohorts (n = 499) further strengthened these findings showing germline carriers had worse OS (p = 0.0009). Additionally, we determined whether tumour mutation burden (TMB) or BRAF status were prognostic markers of survival. Low TMB rate (< 20 Mut/Mb; p = 0.0034) and BRAF p.V600 mutation (p = 0.0355) were associated with worse progression-free survival. Combining these biomarkers indicated that V600 mutant patients had significantly lower TMB (p = 0.0155). This was confirmed in the TCGA (n = 443, p = 0.0007). Integrative analysis showed germline mutation status conferred the highest risk (HR 5.2, 95% CI 1.72–15.7). Stage IV (HR 2.5, 0.74–8.6) and low TMB (HR 2.3, 0.57–9.4) were similar, whereas BRAF V600 status was the weakest prognostic biomarker (HR 1.5, 95% CI 0.44–5.2).
Collapse
Affiliation(s)
- Lauren G Aoude
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| | - Vanessa F Bonazzi
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Sandra Brosda
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Kalpana Patel
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | | | - Harald Oey
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Scott Wood
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - John V Pearson
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - James M Lonie
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Melissa Arneil
- Division of Cancer Services, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| | - Victoria Atkinson
- Queensland Melanoma Project, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland, St Lucia, QLD, 4067, Australia
| | - B Mark Smithers
- Queensland Melanoma Project, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland, St Lucia, QLD, 4067, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Andrew P Barbour
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Queensland Melanoma Project, Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
2334
|
Marsavela G, Lee J, Calapre L, Wong SQ, Pereira MR, McEvoy AC, Reid AL, Robinson C, Warburton L, Abed A, Khattak MA, Meniawy TM, Dawson SJ, Sandhu S, Carlino MS, Menzies AM, Scolyer RA, Long GV, Amanuel B, Millward M, Ziman MR, Rizos H, Gray ES. Circulating Tumor DNA Predicts Outcome from First-, but not Second-line Treatment and Identifies Melanoma Patients Who May Benefit from Combination Immunotherapy. Clin Cancer Res 2020; 26:5926-5933. [PMID: 33067256 DOI: 10.1158/1078-0432.ccr-20-2251] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/29/2020] [Accepted: 09/09/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE We evaluated the predictive value of pretreatment ctDNA to inform therapeutic outcomes in patients with metastatic melanoma relative to type and line of treatment. EXPERIMENTAL DESIGN Plasma circulating tumor DNA (ctDNA) was quantified in 125 samples collected from 110 patients prior to commencing treatment with immune checkpoint inhibitors (ICIs), as first- (n = 32) or second-line (n = 27) regimens, or prior to commencing first-line BRAF/MEK inhibitor therapy (n = 66). An external validation cohort included 128 patients commencing ICI therapies in the first- (N = 77) or second-line (N = 51) settings. RESULTS In the discovery cohort, low ctDNA (≤20 copies/mL) prior to commencing therapy predicted longer progression-free survival (PFS) in patients treated with first-line ICIs [HR, 0.20; 95% confidence interval (CI) 0.07-0.53; P < 0.0001], but not in the second-line setting. An independent cohort validated that ctDNA is predictive of PFS in the first-line setting (HR, 0.42; 95% CI, 0.22-0.83; P = 0.006), but not in the second-line ICI setting. Moreover, ctDNA prior to commencing ICI treatment was not predictive of PFS for patients pretreated with BRAF/MEK inhibitors in either the discovery or validation cohorts. Reduced PFS and overall survival were observed in patients with high ctDNA receiving anti-PD-1 monotherapy, relative to those treated with combination anti-CTLA-4/anti-PD-1 inhibitors. CONCLUSIONS Pretreatment ctDNA is a reliable indicator of patient outcome in the first-line ICI treatment setting, but not in the second-line ICI setting, especially in patients pretreated with BRAF/MEK inhibitors. Preliminary evidence indicated that treatment-naïve patients with high ctDNA may preferentially benefit from combined ICIs.
Collapse
Affiliation(s)
- Gabriela Marsavela
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Jenny Lee
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Leslie Calapre
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Centre, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Michelle R Pereira
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Ashleigh C McEvoy
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Anna L Reid
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Cleo Robinson
- Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Australia.,School of Biomedical Science, University of Western Australia, Crawley, Australia
| | - Lydia Warburton
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Afaf Abed
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia
| | - Muhammad A Khattak
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia.,School of Medicine, University of Western Australia, Crawley, Australia
| | - Tarek M Meniawy
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,School of Medicine, University of Western Australia, Crawley, Australia
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and New South Wales Health Pathology, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Benhur Amanuel
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Australia.,School of Medicine, University of Western Australia, Crawley, Australia
| | - Michael Millward
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,School of Medicine, University of Western Australia, Crawley, Australia
| | - Melanie R Ziman
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,School of Biomedical Science, University of Western Australia, Crawley, Australia
| | - Helen Rizos
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.
| |
Collapse
|
2335
|
Kerr SC, Morgan MM, Gillette AA, Livingston MK, Lugo-Cintron KM, Favreau PF, Florek L, Johnson BP, Lang JM, Skala MC, Beebe DJ. A bioengineered organotypic prostate model for the study of tumor microenvironment-induced immune cell activation. Integr Biol (Camb) 2020; 12:250-262. [PMID: 33034643 PMCID: PMC7569006 DOI: 10.1093/intbio/zyaa020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/17/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022]
Abstract
The prostate tumor microenvironment (TME) is strongly immunosuppressive; it is largely driven by alteration in cell phenotypes (i.e. tumor-associated macrophages and exhausted cytotoxic T cells) that result in pro-tumorigenic conditions and tumor growth. A greater understanding into how these altered immune cell phenotypes are developed and could potentially be reversed would provide important insights into improved treatment efficacy for prostate cancer. Here, we report a microfluidic model of the prostate TME that mimics prostate ducts across various stages of prostate cancer progression, with associated stroma and immune cells. Using this platform, we exposed immune cells to a benign prostate TME or a metastatic prostate TME and investigated their metabolism, gene and cytokine expression. Immune cells exposed to the metastatic TME showed metabolic differences with a higher redox ratio indicating a switch to a more glycolytic metabolic profile. These cells also increased expression of pro-tumor response cytokines that have been shown to increase cell migration and angiogenesis such as Interleukin-1 (IL-1) a and Granulocyte-macrophage colony-stimulating factor (GM-CSF). Lastly, we observed decreased TLR, STAT signaling and TRAIL expression, suggesting that phenotypes derived from exposure to the metastatic TME could have an impaired anti-tumor response. This platform could provide a valuable tool for studying immune cell phenotypes in in vitro tumor microenvironments.
Collapse
Affiliation(s)
- Sheena C Kerr
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Molly M Morgan
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Amani A Gillette
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Megan K Livingston
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Karina M Lugo-Cintron
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Logan Florek
- Morgridge Institute for Research, Madison, WI, USA
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Melissa C Skala
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2336
|
Siciliano MA, Dastoli S, d'Apolito M, Staropoli N, Tassone P, Tagliaferri P, Barbieri V. Pembrolizumab-Induced Psoriasis in Metastatic Melanoma: Activity and Safety of Apremilast, a Case Report. Front Oncol 2020; 10:579445. [PMID: 33163407 PMCID: PMC7591674 DOI: 10.3389/fonc.2020.579445] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/11/2020] [Indexed: 11/13/2022] Open
Abstract
Background Immune checkpoint inhibitors targeting cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), programmed death-1 receptor (PD-1), and programmed death-1 receptor and its ligand (PD-L1) increased the survival of patients affected by metastatic malignant melanoma. Due to their mechanism of action, these drugs are associated with a unique toxicity profile. Indeed, immune-related adverse events (irAEs) present a wide clinical spectrum representing the Achilles' heel of immunotherapy. Overall, cutaneous toxicities are among the most common irAEs. Immunomodulatory drugs are used for the management of irAEs and can theoretically lead to tumor escape. Case Presentation We report the case of a 75-year-old man with metastatic melanoma receiving the anti-PD1 Pembrolizumab therapy. After 10 treatment cycles, the patient came to our clinic with itchy psoriatic manifestations widespread >30% of the body surface [12.3 Psoriasis Area and Severity Index (PASI) score] that negatively impacted on the patient's quality of life and compliance with immunotherapy. Additionally, he had no positive personal history of psoriasis. Given the severity of the cutaneous manifestations, in a multidisciplinary approach, Apremilast (an oral small molecule PDE4 inhibitor) was started. Furthermore, Pembrolizumab was interrupted for 4 weeks until the improvement of skin lesions and the disappearance of itching. Immunosuppressive methylprednisolone therapy was initiated with a dose of 16 mg/die; then, this initial dose was progressively reduced until discontinuation. After 10 months, the patient had a good general clinical condition with psoriasis complete remission. Moreover, positron emission tomography (PET) and computed tomography (CT) scans showed complete response by immune Response Evaluation Criteria in Solid Tumors (iRECIST). Conclusion To the best of our knowledge, this is the first report on the safety and efficacy of Apremilast for the treatment of immunotherapy-induced psoriasis in metastatic melanoma.
Collapse
Affiliation(s)
- Maria Anna Siciliano
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Stefano Dastoli
- Dermatology Unit, Mater Domini Hospital, Catanzaro, Italy.,Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.,Translational Medical Oncology Unit, Mater Domini Hospital, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.,Medical Oncology Unit, Mater Domini Hospital, Catanzaro, Italy
| | - Vito Barbieri
- Medical Oncology Unit, Mater Domini Hospital, Catanzaro, Italy
| |
Collapse
|
2337
|
Cebula H, Noel G, Garnon J, Todeschi J, Burckel H, de Mathelin M, Gangi A, Proust F. The Cryo-immunologic effect: A therapeutic advance in the treatment of glioblastomas? Neurochirurgie 2020; 66:455-460. [PMID: 33045247 DOI: 10.1016/j.neuchi.2020.06.135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/15/2020] [Accepted: 06/28/2020] [Indexed: 11/17/2022]
Abstract
The immunotherapy of cerebral glioblastoma has become a hot topic. Immune checkpoint blockade antibodies have progressively acquired a role in the management of malignant tumors. A multimodal approach using surgery, radiotherapy, chemotherapy in combination with immunotherapy represent a potent weapon against glioblastomas. In parallel, clinical applications of cryotherapy-freezing tumors based on repetition of rapid freeze-slow thaw cycle-for various cancers such as skin, lung, breast, esophagus, hepatic, kidney, prostate and bone tumors were developed. The future immunomodulatory approaches might be combined with brain tumors cryoablation to increase the cryoimmune response. The objective of this study was to analyze from the literature the relationship between cerebral cryosurgery and immunomodulation using PRISMA method. The animals' studies demonstrate the dendritic cells maturation and activation with the enhancement of antigen-presenting function after cryotherapy suggesting the potential usefulness of the association of cryotherapy and immunomodulator in the management of gliomas.
Collapse
Affiliation(s)
- H Cebula
- Department of Neurosurgery, University Hospital of Strasbourg, 67100 Strasbourg, France.
| | - G Noel
- Department of Radiotherapy, ICANS, 67100 Strasbourg, France
| | - J Garnon
- Department of Interventional Radiology, University Hospital of Strasbourg, 67100 Strasbourg, France
| | - J Todeschi
- Department of Neurosurgery, University Hospital of Strasbourg, 67100 Strasbourg, France
| | - H Burckel
- Department of Radiotherapy, ICANS, 67100 Strasbourg, France
| | - M de Mathelin
- Icube-UMR 7357 Télécom Physique, 67100 Strasbourg, France
| | - A Gangi
- Department of Interventional Radiology, University Hospital of Strasbourg, 67100 Strasbourg, France
| | - F Proust
- Department of Neurosurgery, University Hospital of Strasbourg, 67100 Strasbourg, France
| |
Collapse
|
2338
|
Grasso CS, Tsoi J, Onyshchenko M, Abril-Rodriguez G, Ross-Macdonald P, Wind-Rotolo M, Champhekar A, Medina E, Torrejon DY, Shin DS, Tran P, Kim YJ, Puig-Saus C, Campbell K, Vega-Crespo A, Quist M, Martignier C, Luke JJ, Wolchok JD, Johnson DB, Chmielowski B, Hodi FS, Bhatia S, Sharfman W, Urba WJ, Slingluff CL, Diab A, Haanen JBAG, Algarra SM, Pardoll DM, Anagnostou V, Topalian SL, Velculescu VE, Speiser DE, Kalbasi A, Ribas A. Conserved Interferon-γ Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma. Cancer Cell 2020; 38:500-515.e3. [PMID: 32916126 PMCID: PMC7872287 DOI: 10.1016/j.ccell.2020.08.005] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/17/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022]
Abstract
We analyze the transcriptome of baseline and on-therapy tumor biopsies from 101 patients with advanced melanoma treated with nivolumab (anti-PD-1) alone or combined with ipilimumab (anti-CTLA-4). We find that T cell infiltration and interferon-γ (IFN-γ) signaling signatures correspond most highly with clinical response to therapy, with a reciprocal decrease in cell-cycle and WNT signaling pathways in responding biopsies. We model the interaction in 58 human cell lines, where IFN-γ in vitro exposure leads to a conserved transcriptome response unless cells have IFN-γ receptor alterations. This conserved IFN-γ transcriptome response in melanoma cells serves to amplify the antitumor immune response. Therefore, the magnitude of the antitumor T cell response and the corresponding downstream IFN-γ signaling are the main drivers of clinical response or resistance to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Catherine S Grasso
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Jennifer Tsoi
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Mykola Onyshchenko
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Gabriel Abril-Rodriguez
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | | | - Megan Wind-Rotolo
- Translational Bioinformatics, Bristol-Myers Squibb, Hopewell, NJ, USA
| | - Ameya Champhekar
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Egmidio Medina
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Davis Y Torrejon
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Daniel Sanghoon Shin
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Phuong Tran
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Yeon Joo Kim
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Cristina Puig-Saus
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Katie Campbell
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Agustin Vega-Crespo
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Michael Quist
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | | | | | - Jedd D Wolchok
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Bartosz Chmielowski
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - F Stephen Hodi
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Dana Farber Cancer Institute, Boston, MA, USA
| | | | - William Sharfman
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Walter J Urba
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - Adi Diab
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Drew M Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valsamo Anagnostou
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Suzanne L Topalian
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Victor E Velculescu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Anusha Kalbasi
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
2339
|
Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, Mahammedi H, Patnaik A, Subudhi SK, Ciprotti M, Simsek B, Saci A, Hu Y, Han GC, Fizazi K. Nivolumab Plus Ipilimumab for Metastatic Castration-Resistant Prostate Cancer: Preliminary Analysis of Patients in the CheckMate 650 Trial. Cancer Cell 2020; 38:489-499.e3. [PMID: 32916128 DOI: 10.1016/j.ccell.2020.08.007] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/14/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is immunologically "cold" and predominantly resistant to immune checkpoint therapy due to few tumor-infiltrating T cells. Ipilimumab (anti-CTLA-4) or anti-PD-1/PD-L1 monotherapy failed to show a significant benefit. Although the PD-1/PD-L1 pathway is minimally expressed in prostate tumors, we previously demonstrated that PD-1/PD-L1 expression increases as a compensatory inhibitory pathway in parallel with an ipilimumab-induced increase in tumor-infiltrating T cells. Here, we report the largest trial to date in mCRPC with anti-CTLA-4 plus anti-PD-1 (nivolumab 1 mg/kg plus ipilimumab 3 mg/kg; CheckMate 650, NCT02985957). With median follow-ups of 11.9 and 13.5 months in cohorts 1 (pre-chemotherapy; n = 45) and 2 (post-chemotherapy; n = 45), objective response rate was 25% and 10%, and median overall survival was 19.0 and 15.2 months, respectively. Four patients, two in each cohort, had complete responses. Exploratory studies identify potential biomarkers of response. Grade 3-4 treatment-related adverse events have occurred in ∼42%-53% of patients, with four treatment-related deaths. Therefore, dose/schedule modifications have been implemented.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Unit 1374, 1155 Pressler Street, Houston, TX 77030, USA.
| | - Russell K Pachynski
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vivek Narayan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aude Fléchon
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes Aix-Marseille Université, 13009 Marseille, France
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hakim Mahammedi
- Department of Medical Oncology, Centre Jean Perrin, 63011 Clermont-Ferrand, France
| | - Akash Patnaik
- Department of Medical Oncology, The University of Chicago Medicine, Chicago, IL 60637, USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Unit 1374, 1155 Pressler Street, Houston, TX 77030, USA
| | | | | | - Abdel Saci
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | - Yanhua Hu
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | | | - Karim Fizazi
- Department of Cancer Medicine, Gustave Roussy, University of Paris Saclay, 94800 Villejuif, France
| |
Collapse
|
2340
|
Karapetyan L, Luke JJ, Davar D. Toll-Like Receptor 9 Agonists in Cancer. Onco Targets Ther 2020; 13:10039-10060. [PMID: 33116588 PMCID: PMC7553670 DOI: 10.2147/ott.s247050] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/16/2020] [Indexed: 12/19/2022] Open
Abstract
Toll-like receptor 9 (TLR9) is a pattern recognition receptor that is predominantly located intracellularly in immune cells, including dendritic cells, macrophages, natural killer cells, and other antigen-presenting cells (APC). The primary ligands for TLR9 receptors are unmethylated cytidine phosphate guanosine (CpG) oligodinucleotides (ODN). TLR9 agonists induce inflammatory processes that result in the enhanced uptake and killing of microorganisms and cancer cells as well as the generation of adaptive immune responses. Preclinical studies of TLR9 agonists suggested efficacy both as monotherapy and in combination with several agents, which led to clinical trials in patients with advanced cancer. In these studies, intravenous, intratumoral, and subcutaneous routes of administration have been tested; with anti-tumor responses in both treated and untreated metastatic sites. TLR9 agonist monotherapy is safe, although efficacy is minimal in advanced cancer patients; conversely, combinations appear to be more promising. Several ongoing phase I and II clinical trials are evaluating TLR9 agonists in combination with a variety of agents including chemotherapy, radiotherapy, targeted therapy, and immunotherapy agents. In this review article, we describe the distribution, structure and signaling of TLR9; discuss the results of preclinical studies of TLR9 agonists; and review ongoing clinical trials of TLR9 agonists singly and in combination in patients with advanced solid tumors.
Collapse
Affiliation(s)
- Lilit Karapetyan
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Pittsburgh, PA, USA
| | - Jason J Luke
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Pittsburgh, PA, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diwakar Davar
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Pittsburgh, PA, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2341
|
Xu D, Liu H, Xiang M, Feng A, Tian M, Li D, Mao Y, Zhang L, Zhang S, Tian Y. The relationship between pneumonitis and programmed cell death-1/programmed cell death ligand 1 inhibitors among cancer patients: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e22567. [PMID: 33031304 PMCID: PMC7544380 DOI: 10.1097/md.0000000000022567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND We put the meta-analysis into practice to reveal the relationship between the incidence risk of immune-related pneumonitis and the use of programmed cell death-1 (PD-1) and ligand 1 (PD-L1) inhibitors related pneumonitis in cancer patients. METHOD The meta-analysis was put into practice according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Odds ratio (OR) was evaluated by random effect model. RESULTS After screening and eligibility assessment, 33 clinical trials involving 19,854 patients were selected and used for the final meta-analysis after selection criteria checked. Compared with chemotherapy, the use of PD-1/PD-L1 inhibitors alone increased the incidence risk of all-grade (OR = 4.29, 95% confidence interval: [2.97, 6.19], P < .00001) and grade 3 to 5 immune-related pneumonitis (OR = 3.53, 95% confidence interval: [2.04, 6.11], P < .00001). Similar trend could also be found when PD-1/PD-L1 inhibitors were prescribed alone or in combination with other anti-tumor therapies. CONCLUSION Whether PD-1/PD-L1 inhibitors were used alone or combined with other antitumor drugs, the incidence risk of immune-related pneumonitis would be increased.
Collapse
Affiliation(s)
| | - Hongmei Liu
- Department of Radiotherapy Oncology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong
| | - Meiyi Xiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing
| | - Alei Feng
- Department of Oncology, Shandong Provincial Hospital Affiliated with Shandong First Medical University
| | - Mei Tian
- Respiratory Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan
| | - Donghua Li
- Department of Radiotherapy, The People's Hospital of Yuncheng County, Heze
| | - Yantao Mao
- Department of Oncology, Yantaishan Hospital of Shandong Province, Yantai
| | - Li Zhang
- Department of Pathology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong
| | - Shuisheng Zhang
- Department of General Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Yuan Tian
- Department of Radiotherapy Oncology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong
| |
Collapse
|
2342
|
Gkiala A, Palioura S. Conjunctival Melanoma: Update on Genetics, Epigenetics and Targeted Molecular and Immune-Based Therapies. Clin Ophthalmol 2020; 14:3137-3152. [PMID: 33116365 PMCID: PMC7553763 DOI: 10.2147/opth.s271569] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/17/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose To present the molecular mechanisms involved in the pathogenesis of conjunctival melanoma (CM) and review the existing literature on targeted molecular inhibitors as well as immune checkpoint inhibitors for the management of locally advanced and metastatic disease. Methods A comprehensive review of the literature was performed using the keywords "conjunctival melanoma", "immune checkpoint inhibitors", "BRAF inhibitors", "MEK inhibitors", "CTLA4 inhibitors", "PD1 inhibitors", "c-KIT mutations", "BRAF mutations", "NRAS mutations", "dabrafenib", "trametinib", "vemurafenib", "ipilimumab", "pembrolizumab", and "nivolumab". A total of 250 articles were reviewed and 120 were included in this report. Results Mutations of mediators in the MAP kinase pathway, such as RAS, BRAF, MEK and ERK, and mutations of the PI3K/AKT/mTOR pathway play a major role in the pathogenesis of conjunctival melanoma. In addition, alterations of c-KIT, NF1, TERT, chemokine receptors as well as chromosomal copy number alterations and micro RNAs are thought to have a causative association with CM development. Targeted molecular inhibitors, such as BRAF and MEK inhibitors, are currently being implemented in the therapy of BRAF-mutated CM. Furthermore, immune checkpoint PD-1 and CTLA4 inhibitors with favorable clinical outcomes in the treatment of cutaneous melanoma have increased recurrence-free survival and reduced metastatic spread in CM cases. Conclusion The complex molecular mechanisms that contribute to the development of CM can be targeted both by molecular inhibitors of oncogenic pathways as well as immune checkpoint inhibitors in order to halt progression of the disease and increase survival.
Collapse
Affiliation(s)
- Anastasia Gkiala
- National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | | |
Collapse
|
2343
|
Julve M, Clark JJ, Lythgoe MP. Advances in cyclin-dependent kinase inhibitors for the treatment of melanoma. Expert Opin Pharmacother 2020; 22:351-361. [PMID: 33030382 DOI: 10.1080/14656566.2020.1828348] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Despite the recent advances in the treatment of malignant melanoma with immunotherapy and BRAF/MEK targeted agents, advanced disease still beholds a poor prognosis for a significant proportion of patients. Cyclin-dependent kinase (CDK) inhibitors have been investigated as novel melanoma therapeutics throughout a range of phase 1 and 2 trials, as single agents and in combination with established treatments. Areas covered: This article summarizes the rationale for, and development of CDK inhibitors in melanoma, with their evolution from pan-CDK inhibitors to highly specific agents, throughout clinical trials and finally their potential future use. Expert opinion: Whilst CDK inhibitors have been practice changing in breast cancer management, their efficacy is yet to be proven in melanoma. Combination with BRAF/MEK inhibitors has been hindered by dose-limiting toxicities, but their role may yet to be found within the spectrum of biomarker-derived personalized melanoma management. The effect that CDK inhibitors can have as an adjunct to immunotherapy also remains to be seen.
Collapse
Affiliation(s)
- Maximilian Julve
- Department of Surgery & Cancer, Imperial College London , London, UK
| | - James J Clark
- Department of Surgery & Cancer, Imperial College London , London, UK
| | - Mark P Lythgoe
- Department of Surgery & Cancer, Imperial College London , London, UK
| |
Collapse
|
2344
|
Xu Y, Fu Y, Zhu B, Wang J, Zhang B. Predictive Biomarkers of Immune Checkpoint Inhibitors-Related Toxicities. Front Immunol 2020; 11:2023. [PMID: 33123120 PMCID: PMC7572846 DOI: 10.3389/fimmu.2020.02023] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence and continuous development of immune checkpoint inhibitors (ICIs) therapy brings a revolution in cancer therapy history, but the major hurdle associated with their usage is the concomitant ICIs-related toxicities that present a challenge for oncologists. The toxicities may involve non-specific symptoms of multiple systems as for the unique mechanism of formation, which are not easily distinguishable from traditional toxicities. A few of these adverse events are self-limiting and readily manageable, but others may limit treatment, cause interruption and need to be treated with methylprednisolone or tumor necrosis factor-α (TNF-α) antibody infliximab, and even directly threaten life. Early accurate recognition and adequate management are critical to the patient's prognosis and overall survival (OS). Several biomarkers such as the expression of programmed cell death ligand 1 (PD-L1), tumor mutation burden (TMB), and microsatellite instability-high (MSI-H)/mismatch repair-deficient (dMMR) have been proved to be the predictors for anti-tumor efficacy of ICIs, but there is a gap in clinical needs for effective biomarkers that predict toxicities and help filter out the patients who may benefit most from these costly therapies while avoiding major risks of toxicities. Here, we summarize several types of risk factors correlated with ICIs-related toxicities to provide a reference for oncologists to predict the occurrence of ICIs-related toxicities resulting in a timely process in clinical practice.
Collapse
Affiliation(s)
- Ya Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Fu
- Department of Oncology, Xiangyang Hospital, Hubei University of Chinese Medicine, Xiangyang, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Bicheng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2345
|
Tumor Microenvironment: Implications in Melanoma Resistance to Targeted Therapy and Immunotherapy. Cancers (Basel) 2020; 12:cancers12102870. [PMID: 33036192 PMCID: PMC7601592 DOI: 10.3390/cancers12102870] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The response to pharmacological treatments is deeply influenced by the tight interactions between the tumor cells and the microenvironment. In this review we describe, for melanoma, the most important mechanisms of resistance to targeted therapy and immunotherapy mediated by the components of the microenvironment. In addition, we briefly describe the most recent therapeutic advances for this pathology. The knowledge of molecular mechanisms, which are underlying of drug resistance, is fundamental for the development of new therapeutic approaches for the treatment of melanoma patients. Abstract Antitumor therapies have made great strides in recent decades. Chemotherapy, aggressive and unable to discriminate cancer from healthy cells, has given way to personalized treatments that, recognizing and blocking specific molecular targets, have paved the way for targeted and effective therapies. Melanoma was one of the first tumor types to benefit from this new care frontier by introducing specific inhibitors for v-Raf murine sarcoma viral oncogene homolog B (BRAF), mitogen-activated protein kinase kinase (MEK), v-kit Hardy–Zuckerman 4 feline sarcoma viral oncogene homolog (KIT), and, recently, immunotherapy. However, despite the progress made in the melanoma treatment, primary and/or acquired drug resistance remains an unresolved problem. The molecular dynamics that promote this phenomenon are very complex but several studies have shown that the tumor microenvironment (TME) plays, certainly, a key role. In this review, we will describe the new melanoma treatment approaches and we will analyze the mechanisms by which TME promotes resistance to targeted therapy and immunotherapy.
Collapse
|
2346
|
Combined PD-1, BRAF and MEK inhibition in advanced BRAF-mutant melanoma: safety run-in and biomarker cohorts of COMBI-i. Nat Med 2020; 26:1557-1563. [PMID: 33020648 DOI: 10.1038/s41591-020-1082-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
Abstract
Immune and targeted therapies achieve long-term survival in metastatic melanoma; however, new treatment strategies are needed to improve patients' outcomes1,2. We report on the efficacy, safety and biomarker analysis from the single-arm safety run-in (part 1; n = 9) and biomarker (part 2; n = 27) cohorts of the randomized, placebo-controlled, phase 3 COMBI-i trial (NCT02967692) of the anti-PD-1 antibody spartalizumab, in combination with the BRAF inhibitor dabrafenib and MEK inhibitor trametinib. Patients (n = 36) had previously untreated BRAF V600-mutant unresectable or metastatic melanoma. In part 1, the recommended phase 3 regimen was identified based on the incidence of dose-limiting toxicities (DLTs; primary endpoint): 400 mg of spartalizumab every 4 weeks plus 150 mg of dabrafenib twice daily plus 2 mg of trametinib once daily. Part 2 characterized changes in PD-L1 levels and CD8+ cells following treatment (primary endpoint), and analyzed additional biomarkers. Assessments of efficacy and safety were key secondary endpoints (median follow-up, 24.3 months). Spartalizumab plus dabrafenib and trametinib led to an objective response rate (ORR) of 78%, including 44% complete responses (CRs). Grade ≥3 treatment-related adverse events (TRAEs) were experienced by 72% of patients. All patients had temporary dose modifications, and 17% permanently discontinued all three study drugs due to TRAEs. Early progression-free survival (PFS) events were associated with low tumor mutational burden/T cell-inflamed gene expression signature (GES) or high immunosuppressive tumor microenvironment (TME) GES levels at baseline; an immunosuppressive TME may also preclude CR. Overall, the efficacy, safety and on-treatment biomarker modulations associated with spartalizumab plus dabrafenib and trametinib are promising, and biomarkers that may predict long-term benefit were identified.
Collapse
|
2347
|
Drakaki A, Dhillon PK, Wakelee H, Chui SY, Shim J, Kent M, Degaonkar V, Hoang T, McNally V, Luhn P, Gutzmer R. Association of baseline systemic corticosteroid use with overall survival and time to next treatment in patients receiving immune checkpoint inhibitor therapy in real-world US oncology practice for advanced non-small cell lung cancer, melanoma, or urothelial carcinoma. Oncoimmunology 2020; 9:1824645. [PMID: 33101774 PMCID: PMC7553559 DOI: 10.1080/2162402x.2020.1824645] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors (CPIs) have expanded treatment options for patients with solid tumors. Systemic corticosteroids (CSs) have an indispensable role in cancer care, but CS-related immunosuppression may counteract the CPI-driven antitumor immune response. This retrospective study investigated the association between baseline CS use (bCS; ≤14 days before, ≤30 days after CPI initiation) and clinical outcomes in patients with advanced non-small cell lung cancer (aNSCLC), melanoma (aMel), or urothelial carcinoma (aUC). We analyzed data from the Flatiron Health electronic health record-derived de-identified database for adults diagnosed with aNSCLC, aMel, or aUC between January 2011 and June 2017 who received ≥1 CPI monotherapy in any treatment line. Associations of bCS use with overall survival (OS) and time to next treatment (TTNT) were estimated using multivariable Cox proportional hazards models adjusting for demographic and clinical characteristics (i.e., ECOG performance status, site of metastases). In total, 2,213 patients were diagnosed with aNSCLC (n = 862), aMel (n = 742), or aUC (n = 609) and received ≥1 CPI administration. Most patients (67%-95%) received CSs, many during the baseline period (19%-30%). Patients with bCS use had shorter median OS than those with no bCS use for aNSCLC (6.6 vs 10.6 months; P= .00018), aMel (16.4 vs 21.5; P= .095), and aUC (4.1 vs 7.7; P= .0012). bCS use was associated with shorter OS (not significant for aMel) and TTNT in adjusted multivariable analyses, and clinical outcomes were not explained by prior CS use or other measured confounders. These findings suggest a potential association between bCS use and decreased CPI effectiveness, warranting further investigation.
Collapse
Affiliation(s)
- Alexandra Drakaki
- Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Preet K Dhillon
- Personalized Healthcare, Product Developmen, Genentech, Inc, South San Francisco, CA, USA
| | - Heather Wakelee
- Thoracic Oncology, Stanford Medical School, Stanford, CA, USA
| | - Stephen Y Chui
- Product Development Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Jinjoo Shim
- Personalized Healthcare, Product Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthew Kent
- Real World Evidence Solutions, Genesis Research, Hoboken, NJ, USA
| | - Viraj Degaonkar
- Product Development Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Tien Hoang
- Product Development Oncology, Genentech, Inc, South San Francisco, CA, USA
| | - Virginia McNally
- Clinical Development, Roche Products Ltd, Welwyn Garden City, UK
| | - Patricia Luhn
- Personalized Healthcare, Product Developmen, Genentech, Inc, South San Francisco, CA, USA
| | - Ralf Gutzmer
- Clinic for Dermatology, Allergology und Venerology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2348
|
Rodrigues Mantuano N, Natoli M, Zippelius A, Läubli H. Tumor-associated carbohydrates and immunomodulatory lectins as targets for cancer immunotherapy. J Immunother Cancer 2020; 8:jitc-2020-001222. [PMID: 33020245 PMCID: PMC7537339 DOI: 10.1136/jitc-2020-001222] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
During oncogenesis, tumor cells present specific carbohydrate chains that are new targets for cancer immunotherapy. Whereas these tumor-associated carbohydrates (TACA) can be targeted with antibodies and vaccination approaches, TACA including sialic acid-containing glycans are able to inhibit anticancer immune responses by engagement of immune receptors on leukocytes. A family of immune-modulating receptors are sialic acid-binding Siglec receptors that have been recently described to inhibit antitumor activity mediated by myeloid cells, natural killer cells and T cells. Other TACA-binding receptors including selectins have been linked to cancer progression. Recent studies have shown that glycan-lectin interactions can be targeted to improve cancer immunotherapy. For example, interactions between the immune checkpoint T cell immunoglobulin and mucin-domain containing-3 and the lectin galectin-9 are targeted in clinical trials. In addition, an antibody against the lectin Siglec-15 is being tested in an early clinical trial. In this review, we summarize the previous and current efforts to target TACA and to inhibit inhibitory immune receptors binding to TACA including the Siglec-sialoglycan axis.
Collapse
Affiliation(s)
| | - Marina Natoli
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| | - Alfred Zippelius
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| | - Heinz Läubli
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| |
Collapse
|
2349
|
Weilandt J, Diehl K, Schaarschmidt ML, Kiecker F, Sasama B, Pronk M, Ohletz J, Könnecke A, Müller V, Utikal J, Hillen U, Harth W, Peitsch WK. Patient preferences for treatment of advanced melanoma: impact of comorbidities. J Dtsch Dermatol Ges 2020; 19:58-70. [PMID: 33015933 DOI: 10.1111/ddg.14293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/15/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND OBJECTIVES Choice of treatment for advanced melanoma is crucially influenced by comorbidities and patient preferences. Our study aimed to investigate the impact of comorbidities on preferences. PATIENTS AND METHODS 150 patients with melanoma stage IIC-IV completed a discrete choice experiment to determine preferences for outcome (overall response rate [ORR], 2-year survival, progression-free survival [PFS], time to response [TTR], kind of adverse events [AE], AE-related treatment discontinuation) and process attributes (frequency and route of administration [RoA], frequency of consultations) of systemic melanoma treatments. The impact of comorbidities was assessed by analysis of variance and multivariate regression. RESULTS Participants with hypertension and other cardiovascular diseases attached significantly greater importance to TTR and RoA than others. Respondents with arthropathy cared more about TTR (β = 0.179, P = 0.047) and RoA, but less about ORR (β = -0.209, P = 0.021). Individuals with diabetes considered AE (β = 0.185, P = 0.039) and frequency of consultations more essential, but ORR less relevant. Those with other malignancies were particularly worried about treatment discontinuation (β = 0.219, P = 0.008), but less about ORR (β = -0.202, P = 0.015). Participants with depression focused more on PFS (β = 0.201, P = 0.025) and less on TTR (β = -0.201, P = 0.023) and RoA (β = -0.167, P = 0.050). CONCLUSIONS Treatment preferences of melanoma patients vary significantly dependent on comorbidities.
Collapse
Affiliation(s)
- Juliane Weilandt
- Department of Dermatology and Phlebology, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Katharina Diehl
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marthe-Lisa Schaarschmidt
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Felix Kiecker
- Department of Dermatology, Venereology und Allergology, Charité University Medicine Berlin, Berlin, Germany.,Department of Dermatology and Venereology, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Bianca Sasama
- Department of Dermatology and Phlebology, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Melanie Pronk
- Department of Dermatology and Allergology, Vivantes Klinikum Spandau, Berlin, Germany
| | - Jan Ohletz
- Department of Dermatology and Allergology, Vivantes Klinikum Spandau, Berlin, Germany
| | - Andreas Könnecke
- Department of Dermatology and Venereology, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Verena Müller
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany.,Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jochen Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany.,Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Uwe Hillen
- Department of Dermatology and Venereology, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Wolfgang Harth
- Department of Dermatology and Allergology, Vivantes Klinikum Spandau, Berlin, Germany
| | - Wiebke K Peitsch
- Department of Dermatology and Phlebology, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| |
Collapse
|
2350
|
Lamos C, Hunger RE. Checkpoint-Inhibitoren – Indikation und Verwendung bei Melanompatienten. Z Rheumatol 2020; 79:818-825. [PMID: 32926216 PMCID: PMC7550369 DOI: 10.1007/s00393-020-00870-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2020] [Indexed: 01/24/2023]
Abstract
ZusammenfassungDie gesamte Onkologie wurde durch die Einführung von Ipilimumab, einem Checkpoint-Inhibitor, im Jahr 2011 revolutioniert. Seitdem wurden weitere effektive Checkpoint-Inhibitoren, wie die PD-1-Antikörper Nivolumab und Pembrolizumab entwickelt. Die Ergebnisse sind bahnbrechend, insbesondere beim fortgeschrittenen malignen Melanom, welches bis vor Kurzem in den meisten Fällen nach wenigen Monaten zum Tode führte. Die Anwendung der Checkpoint-Inhibitoren wurde mit vielversprechenden Ergebnissen auf weitere Tumorentitäten ausgeweitet.
Collapse
Affiliation(s)
- C Lamos
- Universitätsklinik für Dermatologie, Inselspital Bern, Freiburgstr. 34, 3010, Bern, Schweiz.
| | - R E Hunger
- Universitätsklinik für Dermatologie, Inselspital Bern, Freiburgstr. 34, 3010, Bern, Schweiz
| |
Collapse
|