1
|
Grice S, Olsson-Brown A, Naisbitt DJ, Hammond S. Immunological Drug-Drug Interactions Affect the Efficacy and Safety of Immune Checkpoint Inhibitor Therapies. Chem Res Toxicol 2024; 37:1086-1103. [PMID: 38912648 PMCID: PMC11256900 DOI: 10.1021/acs.chemrestox.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024]
Abstract
With the rapid expansion in the development and clinical utility of immune checkpoint inhibitors (ICIs) for oncology, the continual evaluation of the safety profile of such agents is imperative. The safety profile of ICIs as monotherapy is dominated by immune-related adverse events, which can be considered as an extension of the mechanism of action of these immunomodulatory drugs. Further to this, an emerging theme is that ICI treatment can significantly impact upon the tolerability of coadministered medications. Numerous reports in literature indicate that ICIs may alter the immunological perception of coadministered drugs, resulting in undesirable reactions to a variety of concomitant medications. These reactions can be severe in manifestation, including hepatotoxicity and Stevens-Johnson Syndrome (SJS)/toxic epidermal necrolysis (TEN), but may also have detrimental impact on malignancy control. To minimize the impact of such drug-drug interactions on patients, it is imperative to identify medications that may cause these reactions, understand the underlying mechanisms, consider the timing and dosing of comedication, and explore alternative medications with comparable efficacies. Improving our understanding of how concomitant medications affect the safety and efficacy of ICIs can allow for potential culprit drugs to be identified/removed/desensitized. This approach will allow the continuation of ICI therapy that may have been discontinued otherwise, thereby improving malignant control and patient and drug development outcomes.
Collapse
Affiliation(s)
- Sophie Grice
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Anna Olsson-Brown
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
- Sussex
Cancer Centre, University Hospitals Sussex, Brighton BN2 5BD, U.K.
| | - Dean J. Naisbitt
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Sean Hammond
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
- ApconiX, Alderley Edge SK10 4TG, U.K.
| |
Collapse
|
2
|
Hornsteiner F, Vierthaler J, Strandt H, Resag A, Fu Z, Ausserhofer M, Tripp CH, Dieckmann S, Kanduth M, Farrand K, Bregar S, Nemati N, Hermann-Kleiter N, Seretis A, Morla S, Mullins D, Finotello F, Trajanoski Z, Wollmann G, Ronchese F, Schmitz M, Hermans IF, Stoitzner P. Tumor-targeted therapy with BRAF-inhibitor recruits activated dendritic cells to promote tumor immunity in melanoma. J Immunother Cancer 2024; 12:e008606. [PMID: 38631706 PMCID: PMC11029477 DOI: 10.1136/jitc-2023-008606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Tumor-targeted therapy causes impressive tumor regression, but the emergence of resistance limits long-term survival benefits in patients. Little information is available on the role of the myeloid cell network, especially dendritic cells (DC) during tumor-targeted therapy. METHODS Here, we investigated therapy-mediated immunological alterations in the tumor microenvironment (TME) and tumor-draining lymph nodes (LN) in the D4M.3A preclinical melanoma mouse model (harboring the V-Raf murine sarcoma viral oncogene homolog B (BRAF)V600E mutation) by using high-dimensional multicolor flow cytometry in combination with multiplex immunohistochemistry. This was complemented with RNA sequencing and cytokine quantification to characterize the immune status of the tumors. The importance of T cells during tumor-targeted therapy was investigated by depleting CD4+ or CD8+ T cells in tumor-bearing mice. Tumor antigen-specific T-cell responses were characterized by performing in vivo T-cell proliferation assays and the contribution of conventional type 1 DC (cDC1) to T-cell immunity during tumor-targeted therapy was assessed using Batf3-/- mice lacking cDC1. RESULTS Our findings reveal that BRAF-inhibitor therapy increased tumor immunogenicity, reflected by an upregulation of genes associated with immune activation. The T cell-inflamed TME contained higher numbers of activated cDC1 and cDC2 but also inflammatory CCR2-expressing monocytes. At the same time, tumor-targeted therapy enhanced the frequency of migratory, activated DC subsets in tumor-draining LN. Even more, we identified a cDC2 population expressing the Fc gamma receptor I (FcγRI)/CD64 in tumors and LN that displayed high levels of CD40 and CCR7 indicating involvement in T cell-mediated tumor immunity. The importance of cDC2 is underlined by just a partial loss of therapy response in a cDC1-deficient mouse model. Both CD4+ and CD8+ T cells were essential for therapy response as their respective depletion impaired therapy success. On resistance development, the tumors reverted to an immunologically inert state with a loss of DC and inflammatory monocytes together with the accumulation of regulatory T cells. Moreover, tumor antigen-specific CD8+ T cells were compromised in proliferation and interferon-γ-production. CONCLUSION Our results give novel insights into the remodeling of the myeloid landscape by tumor-targeted therapy. We demonstrate that the transient immunogenic tumor milieu contains more activated DC. This knowledge has important implications for the development of future combinatorial therapies.
Collapse
Affiliation(s)
- Florian Hornsteiner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Vierthaler
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helen Strandt
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Antonia Resag
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Zhe Fu
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Markus Ausserhofer
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sophie Dieckmann
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Kanduth
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kathryn Farrand
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sarah Bregar
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Niloofar Nemati
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Athanasios Seretis
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Sudhir Morla
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - David Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Francesca Finotello
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, Innsbruck, Austria
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Guido Wollmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
3
|
Rehman S, Venna P, Davis S, Gopagoni R, Uttam R, Farrukh AM, Salehi M. Primary gallbladder melanoma: A systematic review of literature. Ann Diagn Pathol 2024; 68:152244. [PMID: 38103326 DOI: 10.1016/j.anndiagpath.2023.152244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Primary gallbladder melanoma (PGM) is a rare malignancy with only sporadic cases reported in the English literature. We performed a systematic review of the cases published in the PubMed, Science Direct and Google Scholar databases with the aim of describing the reported clinicopathologic features of PGM. Thirty-six articles reporting on 39 patients were reviewed. There was a male predominance, with 23 (64 %) of 36 patients being males. The mean age at presentation was 55 ±16 years. Pain in the right upper quadrant was reported in 20/27 (74 %). The average size of the tumor was 3.5 × 1.9 × 1.4 cm. Gallbladder calculi were reported in 7/27 (26 %). A cholecystectomy was performed in 34/38 (89.5 %). Grossly, the tumor mostly (96.5 %) had polypoid appearances and on microscopic examination, the tumor were predominantly comprised of epithelioid cells 12/17 (70.6 %). Mitotic figures and prominent nucleoli were reportedly found in 8/8 (100 %) and 3/3 (100 %) respectively. Junctional melanocytic components were present in 13/21 (61.9 %). Tumor cells were reportedly immunoreactive for S-100 and HMB-45 in all tested cases. Metastasis were reported in 25/36 (69.4 %), with lymph nodes being the most common site (n = 8), followed by brain (n = 6) and liver (n = 4) for metastasis. At a mean follow-up period of 19 +/- 3 months, 16 (48.5 %) of the 33 patients with available survival data were alive and 17/33 (51.5 %) were dead of disease. There is a lack of unified criteria for the diagnosis of PGM, and future studies should aim to resolve this.
Collapse
Affiliation(s)
| | | | | | | | - Ritika Uttam
- JJM Medical College, Davangere, Karnataka, India
| | | | | |
Collapse
|
4
|
Bashari N, Safaei Lari M, Darvishi A, Daroudi R. Cost-utility analysis of Pembrolizumab compared to other alternative immunotherapy and chemotherapy treatments for patients with advanced melanoma in Iran. Expert Rev Pharmacoecon Outcomes Res 2024; 24:273-284. [PMID: 37750606 DOI: 10.1080/14737167.2023.2263164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVES Immunotherapy drugs like Pembrolizumab have shown significant improvements in treatment outcomes of advanced melanoma. This study aimed to evaluate the cost-utility of Pembrolizumab compared to other immunotherapy and chemotherapy drugs in the first-line treatment of advanced melanoma in Iran. METHODS A partitioned-survival model, based on data from a recent randomized phase 3 study (KEYNOTE-006) and recent meta-analysis, was used to divide Overall survival (OS) time into Progression-free survival (PFS) and post-progression survival for Pembrolizumab, Nivolumab, Ipilimumab, Dacarbazine, Temozolomide, Carboplatin, and Paclitaxel combination. Quality Life Years (QALY) and Incremental Cost-Effectiveness Ratio (ICER) were considered as the final outcome. RESULTS The ICER of Ipilimumab, Nivolumab, Nivolumab & Ipilimumab, and Pembrolizumab compared to Temozolomide was calculated as $40,365.53, $19,591.13, $24,578, and $47,324.2 per QALY, respectively. Scenario analysis demonstrated if the price of one vial of Nivolumab 100 is $90.51, each vial of Pembrolizumab is $119.20, and each vial of Ipilimumab is $101.54, they will be cost-effective in Iran. CONCLUSION None of the immunotherapy drugs studied were found to be cost-effective when considering the cost-effectiveness threshold of $3,532. Therefore, a cost reduction of more than 90% in the prices of immunotherapy drugs would be necessary for them to be considered cost-effective in Iran.
Collapse
Affiliation(s)
- Negin Bashari
- National Center for Health Insurance Research, Tehran, Iran
- Department of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Safaei Lari
- Department of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Darvishi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rajabali Daroudi
- National Center for Health Insurance Research, Tehran, Iran
- Department of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Zhao N, Jiang A, Shang X, Zhao F, Wang R, Fu X, Ruan Z, Liang X, Tian T, Yao Y, Li C. Construction and Evaluation of Clinical Prediction Model for Immunotherapy-related Adverse Events and Clinical Benefit in Cancer Patients Receiving Immune Checkpoint Inhibitors Based on Serum Cytokine Levels. J Immunother 2023; 46:310-322. [PMID: 37335173 PMCID: PMC10473032 DOI: 10.1097/cji.0000000000000478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/27/2023] [Indexed: 06/21/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the therapeutic landscape of cancer therapy. This study aimed to develop novel risk classifiers to predict the risk of immune-related adverse events (irAEs) and the probability of clinical benefits. Patients with cancer who received ICIs from the First Affiliated Hospital of Xi 'an Jiaotong University from November 2020 to October 2022 were recruited and followed up. Logistic regression analyses were performed to identify independent predictive factors for irAEs and clinical response. Two nomograms were developed to predict the irAEs and clinical responses of these individuals, with a receiver operating characteristic curve to assess their predictive ability. Decision curve analysis was performed to estimate the clinical utility of the nomogram. This study included 583 patients with cancer. Among them, 111 (19.0%) developed irAEs. Duration of treatment (DOT)>3 cycles, hepatic-metastases, IL2>2.225 pg/mL, and IL8>7.39 pg/mL were correlated with higher irAEs risk. A total of 347 patients were included in the final efficacy analysis, with an overall clinical benefit rate of 39.7%. DOT>3 cycles, nonhepatic-metastases, and irAEs and IL8>7.39 pg/mL were independent predictive factors of clinical benefit. Ultimately, 2 nomograms were successfully established to predict the probability of irAEs and their clinical benefits. Ultimately, 2 nomograms were successfully established to predict the probability of irAEs and clinical benefits. The receiver operating characteristic curves yielded acceptable nomogram performance. Calibration curves and decision curve analysis supported the hypothesis that nomograms could provide more significant net clinical benefits to these patients. Specific baseline plasma cytokines were closely correlated with irAEs and clinical responses in these individuals.
Collapse
|
6
|
Zhang MJ, Liang MY, Yang SC, Ma XB, Wan SC, Yang QC, Wang S, Xu Z, Sun ZJ. Bioengineering of BRAF and COX2 inhibitor nanogels to boost the immunotherapy of melanoma via pyroptosis. Chem Commun (Camb) 2023; 59:932-935. [PMID: 36597866 DOI: 10.1039/d2cc05498a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glutathione-responsive nanogels (CDNPs) crosslinked via crosslinker DBHD with the BRAF inhibitor dabrafenib and the COX2 inhibitor celecoxib were fabricated. The CDNPs can effectively induce tumor cell pyroptosis to activate robust antitumor immunity. Additionally, CDNPs combined with αPD-1 antibody greatly inhibited tumor growth in a melanoma mouse model with a prolonged survival time.
Collapse
Affiliation(s)
- Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China.
| | - Meng-Yun Liang
- School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China.
| | - Shao-Chen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China.
| | - Xian-Bin Ma
- School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China.
| | - Shu-Cheng Wan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China.
| | - Qi-Chao Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China.
| | - Shuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China.
| | - Zhigang Xu
- School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, P. R. China.
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China.
| |
Collapse
|
7
|
Yin N, Liu X, Ye X, Song W, Lu J, Chen X. PD-1 inhibitor therapy causes multisystem immune adverse reactions: a case report and literature review. Front Oncol 2022; 12:961266. [PMID: 36119464 PMCID: PMC9478917 DOI: 10.3389/fonc.2022.961266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors(ICIs), including cytotoxic T-lymphocyte antigen 4 (anti-CTLA-4), programmed cell death protein 1 and its ligand (PD-1/PD-L1) inhibitors, have been shown to have antitumor activity in various solid tumors. Their mechanism of action is to selectively restore and normalize the body’s immune reponses by disrupting the immunosuppressive signals mediated by PD-1, PD-L1 and CTLA-4 in the tumor microenvironment. With the increase in clinical applications of ICIs, reports of immune-related adverse events (irAEs) have also increased. This article reports a case of a lung cancer patient who developed multisystemic adverse effects after PD-1 inhibitor application: myocarditis, myositis and thrombocytopenia, and analyzes the role of Interleukin 6(IL-6)in the management of irAEs. Despite the patient’s eventual discontinuation of antitumor therapy due to severe irAEs, a significant and durable therapeutic response was observed.
Collapse
|
8
|
Ohanna M, Biber P, Deckert M. Emerging Role of Deubiquitinating Enzymes (DUBs) in Melanoma Pathogenesis. Cancers (Basel) 2022; 14:3371. [PMID: 35884430 PMCID: PMC9322030 DOI: 10.3390/cancers14143371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Metastatic melanoma is the leading cause of death from skin cancer. Therapies targeting the BRAF oncogenic pathway and immunotherapies show remarkable clinical efficacy. However, these treatments are limited to subgroups of patients and relapse is common. Overall, the majority of patients require additional treatments, justifying the development of new therapeutic strategies. Non-genetic and genetic alterations are considered to be important drivers of cellular adaptation mechanisms to current therapies and disease relapse. Importantly, modification of the overall proteome in response to non-genetic and genetic events supports major cellular changes that are required for the survival, proliferation, and migration of melanoma cells. However, the mechanisms underlying these adaptive responses remain to be investigated. The major contributor to proteome remodeling involves the ubiquitin pathway, ubiquitinating enzymes, and ubiquitin-specific proteases also known as DeUBiquitinases (DUBs). In this review, we summarize the current knowledge regarding the nature and roles of the DUBs recently identified in melanoma progression and therapeutic resistance and discuss their potential as novel sources of vulnerability for melanoma therapy.
Collapse
Affiliation(s)
- Mickael Ohanna
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| | - Pierric Biber
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| | - Marcel Deckert
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France; (P.B.); (M.D.)
- Team MicroCan, Equipe Labellisée Ligue Contre le Cancer, 06204 Nice, France
| |
Collapse
|
9
|
Emerging concepts in designing next-generation multifunctional nanomedicine for cancer treatment. Biosci Rep 2022; 42:231373. [PMID: 35638450 PMCID: PMC9272595 DOI: 10.1042/bsr20212051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Nanotherapy has emerged as an improved anticancer therapeutic strategy to circumvent the harmful side effects of chemotherapy. It has been proven to be beneficial to offer multiple advantages, including their capacity to carry different therapeutic agents, longer circulation time and increased therapeutic index with reduced toxicity. Over time, nanotherapy evolved in terms of their designing strategies like geometry, size, composition or chemistry to circumvent the biological barriers. Multifunctional nanoscale materials are widely used as molecular transporter for delivering therapeutics and imaging agents. Nanomedicine involving multi-component chemotherapeutic drug-based combination therapy has been found to be an improved promising approach to increase the efficacy of cancer treatment. Next-generation nanomedicine has also utilized and combined immunotherapy to increase its therapeutic efficacy. It helps in targeting tumor immune response sparing the healthy systemic immune function. In this review, we have summarized the progress of nanotechnology in terms of nanoparticle designing and targeting cancer. We have also discussed its further applications in combination therapy and cancer immunotherapy. Integrating patient-specific proteomics and biomarker based information and harnessing clinically safe nanotechnology, the development of precision nanomedicine could revolutionize the effective cancer therapy.
Collapse
|
10
|
Hu T, Sun W, Xu Y, Qu X, Jin Y, Luo Z, Chen Y. Combination of pembrolizumab plus temozolomide therapy in unresectable and advanced melanoma: a multicenter retrospective analysis in China. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1625. [PMID: 34926669 PMCID: PMC8640901 DOI: 10.21037/atm-21-5738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/10/2021] [Indexed: 11/06/2022]
Abstract
Background This study aimed to evaluate the effect of anti-PD-1 combined with temozolomide as front-line therapy in patients with unresectable advanced melanoma. Methods The records of patients with unresectable advanced melanoma first treated with pembrolizumab plus temozolomide, pembrolizumab alone, or temozolomide-based chemotherapy at three cancer centers from May 2018 to February 2020 were reviewed. Patients were followed up until death or October 30, 2020. Data were retrospectively reviewed and statistically analyzed for the best objective response rate (ORR) and progression-free survival (PFS), as well as toxicities. Results Sixty-nine individuals were identified, including 28 (40.6%) with acral melanoma, 18 (26.1%) with cutaneous melanoma, 21 (30.4%) with mucosal melanoma, and two (2.9%) with unknown primary melanoma. The ORR of pembrolizumab plus temozolomide (8/20, 40.0%) in advanced melanoma was higher than pembrolizumab (3/24, 12.5%) and chemotherapy (1/25, 4.0%) alone as front-line therapies. The median PFS of pembrolizumab plus temozolomide as front-line therapy for advanced melanoma was 9.8 months [95% confidence interval (CI): 1.7-17.9 months], which was a significant improvement on the chemotherapy PFS of 4.2 months (95% CI: 2.6-5.8 months) [hazard ratio (HR) 0.415, 95% CI: 0.185-0.931, P=0.033]. The median PFS of pembrolizumab was 6.2 months (95% CI: 2.5-9.9), with no significant difference compared with chemotherapy (HR 0.647, 95% CI: 0.334-1.252, P=0.196). Conclusions Combining anti-PD-1 with temozolomide has better efficacy than temozolomide-based chemotherapy or anti-PD-1 alone for advanced melanoma treatment without increasing toxicity. Therefore, anti-PD-1 combined with temozolomide may be preferentially used as a front-line regimen for unresectable advanced melanoma.
Collapse
Affiliation(s)
- Tu Hu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Sun
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Xu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinglong Qu
- Department of Surgical Oncology, Fudan University Shanghai Cancer Center Minhang Branch, Shanghai, China
| | - Yongjia Jin
- Department of Orthopedics, Shanghai Electric Power Hospital, Shanghai, China
| | - Zhiguo Luo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yong Chen
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Idel C, Ribbat-Idel J, Klapper L, Krupar R, Bruchhage KL, Dreyer E, Rades D, Polasky C, Offermann A, Kirfel J, Perner S, Wollenberg B. Spatial Distribution of Immune Cells in Head and Neck Squamous Cell Carcinomas. Front Oncol 2021; 11:712788. [PMID: 34778030 PMCID: PMC8581660 DOI: 10.3389/fonc.2021.712788] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background Head and neck squamous cell carcinomas (HNSCCs) have a very moderate response rate to immune checkpoint inhibitor (ICI) treatment compared to other cancer types. Lacking predictive markers for treatment response, we analyzed the immune status of HNSCC and assessed the spatial distribution of immune cells. Materials and Methods Via assessing hematoxylin–eosin (H&E) stains, we divided HNSCCs by the immune cell distribution in hot, cold, and excluded tumors. For each group, each with 10 tumors, we performed serial immunohistochemical (IHC) staining of the immune cell markers, checkpoint molecules, and immune regulators. Results The spatial distributions were different for each immune cell type, allocating regulatory T cells (Tregs) and CD11b cells predominantly in the stroma. CD4 and CD8 cells were present either in the tumor stroma or between cancer cells. Interestingly, the expressions of PD-1 (programmed cell death 1 receptor) and PD-L1 (programmed death-ligand 1) were higher in hot tumors in comparison to cold and excluded tumors. The expression of pSMAD [indicating active transforming growth factor beta (TGF-β)] was higher in excluded tumors. Conclusion Different immune cell distribution patterns within tumors might be crucial for ICI treatment response since hot tumors have the highest expressions of PD-1 and PD-L1. TGF-β might be a key regulator for immune cell distribution and a promising therapeutic target that determines the formation of hot or excluded immune patterns.
Collapse
Affiliation(s)
- Christian Idel
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| | - Julika Ribbat-Idel
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Luise Klapper
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Rosemarie Krupar
- Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Eva Dreyer
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University of Luebeck, Lübeck, Germany
| | - Christina Polasky
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| | - Anne Offermann
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Jutta Kirfel
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology, MRI Technical University Munich, Munich, Germany
| |
Collapse
|
12
|
Interleukin-6 blockade for prophylaxis and management of immune-related adverse events in cancer immunotherapy. Eur J Cancer 2021; 157:214-224. [PMID: 34536945 DOI: 10.1016/j.ejca.2021.08.031] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/15/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have activity across many tumor types, but activation of the immune system may also lead to significant, often steroid-refractory immune-related adverse events (irAEs). We sought to determine the activity of tocilizumab, an anti-interleukin-6 receptor monoclonal antibody, in treatment or prevention of auto-immune irAE in ICI-treated patients. METHODS Institutional databases from 2 melanoma centers were reviewed for patients treated with ICIs and tocilizumab. Longitudinal assessment of C-reactive protein (CRP) and assessment of clinical improvement or prevention of flare of pre-existing auto-immune conditions were utilised to evaluate the benefit of tocilizumab. RESULTS Twenty-two patients were identified. Two were treated prophylactically. Twenty were treated for management of irAEs. Median time to irAE onset from ICI start was 48 days (range 8-786) and from irAE onset to tocilizumab 32 days (range 1-192). Median time to irAE resolution from tocilizumab was 6.5 days (range 1-93). Clinical improvement/benefit was demonstrated in 21/22 patients. Median CRP prior to ICI administration was 32 mg/l (range 0.3-99), at the onset of irAE 49.5 mg/L (range 0.3-251, P = 0.047) and after tocilizumab 18 mg/L (range 0.3-18, P = 0.0011). Tocilizumab was well tolerated with self-limiting and transient toxicities in 11 (50%) patients. From start of ICI, median progression-free survival was 6 months (range 3.9-18.8) and median overall survival was not reached. CONCLUSIONS Tocilizumab was a well-tolerated and effective steroid-sparing treatment for both management of irAEs, as well as prevention of flare of pre-existing auto-immune disorders. Prospective trials to evaluate its efficacy and impact on cancer outcomes compared with standard strategies are required.
Collapse
|
13
|
Sabbah M, Najem A, Krayem M, Awada A, Journe F, Ghanem GE. RTK Inhibitors in Melanoma: From Bench to Bedside. Cancers (Basel) 2021; 13:1685. [PMID: 33918490 PMCID: PMC8038208 DOI: 10.3390/cancers13071685] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
MAPK (mitogen activated protein kinase) and PI3K/AKT (Phosphatidylinositol-3-Kinase and Protein Kinase B) pathways play a key role in melanoma progression and metastasis that are regulated by receptor tyrosine kinases (RTKs). Although RTKs are mutated in a small percentage of melanomas, several receptors were found up regulated/altered in various stages of melanoma initiation, progression, or metastasis. Targeting RTKs remains a significant challenge in melanoma, due to their variable expression across different melanoma stages of progression and among melanoma subtypes that consequently affect response to treatment and disease progression. In this review, we discuss in details the activation mechanism of several key RTKs: type III: c-KIT (mast/stem cell growth factor receptor); type I: EGFR (Epidermal growth factor receptor); type VIII: HGFR (hepatocyte growth factor receptor); type V: VEGFR (Vascular endothelial growth factor), structure variants, the function of their structural domains, and their alteration and its association with melanoma initiation and progression. Furthermore, several RTK inhibitors targeting the same receptor were tested alone or in combination with other therapies, yielding variable responses among different melanoma groups. Here, we classified RTK inhibitors by families and summarized all tested drugs in melanoma indicating the rationale behind the use of these drugs in each melanoma subgroups from preclinical studies to clinical trials with a specific focus on their purpose of treatment, resulted effect, and outcomes.
Collapse
Affiliation(s)
- Malak Sabbah
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.S.); (A.N.); (M.K.); (F.J.)
| | - Ahmad Najem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.S.); (A.N.); (M.K.); (F.J.)
| | - Mohammad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.S.); (A.N.); (M.K.); (F.J.)
| | - Ahmad Awada
- Medical Oncolgy Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Fabrice Journe
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.S.); (A.N.); (M.K.); (F.J.)
| | - Ghanem E. Ghanem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.S.); (A.N.); (M.K.); (F.J.)
| |
Collapse
|
14
|
Wong HSC, Chang WC. Single-cell melanoma transcriptomes depicting functional versatility and clinical implications of STIM1 in the tumor microenvironment. Am J Cancer Res 2021; 11:5092-5106. [PMID: 33859736 PMCID: PMC8039943 DOI: 10.7150/thno.54134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/06/2020] [Indexed: 12/31/2022] Open
Abstract
Rationale: Previous studies have implicated the functions of stromal interaction molecule 1 (STIM1) in immunity and malignancy, however, the specificity and effects of STIM1 expression in malignant and non-malignant cells in the tumor microenvironment are unclear. Methods: In the current study, we posed two central questions: (1) does STIM1 expression elicit different cellular programs in cell types within the melanoma tumor microenvironment (2) whether the expression of STIM1 and STIM1-coexpressed genes (SCGs) serve as prognostic indicators of patient's outcomes? To answer these questions, we dissected cell-specific STIM1-associated cellular programs in diverse cell types within the melanoma tumor microenvironment by measuring cell-type specificity of STIM1 expression and SCGs. Results: A distinct set of SCGs was highly affected in malignant melanoma cells, but not in the other cell types, suggesting the existence of malignant-cell-specific cellular programs reflected by STIM1 expression. In contrast to malignant cells, STIM1 expression appeared to trigger universal and non-specific biological functions in non-malignant cell types, as exemplified by the transcriptomes of macrophages and CD4+ T regulatory cells. Results from bioinformatic analyses indicated that SCGs in malignant cells may alter cell-cell interactions through cytokine/chemokine signaling and/or orchestrate immune infiltration into the tumor. Moreover, a prognostic association between SCGs in CD4+ T regulatory cells and patient's outcomes was identified. However, we didn't find any correlation between SCGs and responsiveness of immunotherapy. Conclusions: Overall, our results provide an integrated biological framework for understanding the functional and clinical consequences of cell-specific STIM1 expression in melanoma.
Collapse
|
15
|
Zhang H, Wang Y, Zheng Q, Tang K, Fang R, Wang Y, Sun Q. Research Interest and Public Interest in Melanoma: A Bibliometric and Google Trends Analysis. Front Oncol 2021; 11:629687. [PMID: 33680968 PMCID: PMC7930473 DOI: 10.3389/fonc.2021.629687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/04/2021] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Melanoma is a severe skin cancer that metastasizes quickly. Bibliometric analysis can quantify hotspots of research interest. Google Trends can provide information to address public concerns. METHODS The top 15 most frequently cited articles on melanoma each year from 2015 to 2019, according to annual citations, were retrieved from the Web of Science database. Original articles, reviews, and research letters were included in this research. For the Google Trends analysis, the topic "Melanoma" was selected as the keyword. Online search data from 2004 to 2019 were collected. Four countries (New Zealand, Australia, the United States and the United Kingdom) were selected for seasonal analysis. Annual trends in relative search volume and seasonal variation were analyzed, and the top related topics and rising related topics were also selected and analyzed. RESULTS The top 15 most frequently cited articles each year were all original articles that focused on immunotherapy (n=8), omics (n=5), and the microbiome (n=2). The average relative search volume remained relatively stable across the years. The seasonal variation analysis revealed that the peak appeared in summer, and the valley appeared in winter. The diseases associated with or manifestations of melanoma, treatment options, risk factors, diagnostic tools, and prognosis were the topics in which the public was most interested. Most of the topics revealed by bibliometric and Google Trends analyses were consistent, with the exception of issues related to the molecular biology of melanoma. CONCLUSION This study revealed the trends in research interest and public interest in melanoma, which may pave the way for further research.
Collapse
|
16
|
Ribbat-Idel J, Perner S, Kuppler P, Klapper L, Krupar R, Watermann C, Paulsen FO, Offermann A, Bruchhage KL, Wollenberg B, Idel C. Immunologic "Cold" Squamous Cell Carcinomas of the Head and Neck Are Associated With an Unfavorable Prognosis. Front Med (Lausanne) 2021; 8:622330. [PMID: 33585526 PMCID: PMC7873597 DOI: 10.3389/fmed.2021.622330] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Head and neck squamous cell carcinoma (HNSCC) represents a common cancer worldwide. Past therapeutic advances have not significantly improved HNSCC prognosis. Therefore, it is necessary to further stratify HNSCC, especially with recent advances in tumor immunology. Methods: Tissue microarrays were assembled from tumor tissue samples and were complemented with comprehensive clinicopathological data of n = 419 patients. H&E whole slides from resection specimen (n = 289) were categorized according to their immune cell infiltrate as “hot,” “cold,” or “excluded.” Results: Investigating tumor immune cell patterns, we found significant differences in survival rates. Immunologic “hot” and “excluded” HNSCCs are associated with better overall survival than “cold” HNSCC patients (p < 0.05). Interestingly, the percentage of all three patterns is nearly identical in p16 positive and negative HNSCCs. Conclusions: Using a plain histological H&E approach to categorize HNSCC as being immunologic “hot,” “cold,” or “excluded” can offer a forecast of patients' prognosis and may thus aid as a potential prognostic tool in routine pathology reports. This “hot-cold-excluded” scheme needs to be applied to more HNSCC cohorts and possibly to other cancer types to determine prognostic meaning, e.g., regarding OS or DFS. Furthermore, our cohort reflects epidemiological data in the national, European, and international context. It may, therefore, be of use for future HNSCC characterization.
Collapse
Affiliation(s)
- Julika Ribbat-Idel
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Patrick Kuppler
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Luise Klapper
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Rosemarie Krupar
- Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Christian Watermann
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Finn-Ole Paulsen
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Anne Offermann
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | | | - Barbara Wollenberg
- Department of Otorhinolaryngology, München rechts der Isar Technical University Munich, Munich, Germany
| | - Christian Idel
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| |
Collapse
|
17
|
Byeon S, Cho HJ, Jang KT, Kwon M, Lee J, Lee J, Kim ST. Molecular profiling of Asian patients with advanced melanoma receiving check-point inhibitor treatment. ESMO Open 2020; 6:100002. [PMID: 33399091 PMCID: PMC7910729 DOI: 10.1016/j.esmoop.2020.100002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/21/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Melanoma is major medical challenge and being able to monitor treatment response is critical. This study aimed to use molecular profiling of Asian patients with advanced melanoma who were receiving treatment with check-point inhibitors (CPIs) to identify novel biomarkers of tumor response. Methods Next-generation sequencing (NGS) was performed using tumor specimens collected from 178 Asian patients with metastatic melanoma receiving CPIs. The NGS data and clinical-pathological factors were analyzed for potential genetic biomarkers of tumor response to CPI treatment. Results The most common melanoma subtype was acral melanoma (40%), followed by cutaneous melanoma (32%), mucosal melanoma (26%), and others (2%). For calculation of treatment efficacy, 164 of the patients could be evaluated. The overall response rate was 45.7%, of which 41 cases exhibited complete responses (25.0%) and 34 showed partial responses (20.7%). There were no significant differences in tumor responses based on melanoma subtype (P = 0.295). Genetically, NRAS mutations, TP53 mutations, and NF2 deletions were significantly associated with resistance to CPIs (P < 0.05). In contrast, MYC and RPS6KB1 amplifications were associated with responsiveness to CPIs (P < 0.05). Median progression-free survival (PFS) for patients treated with CPIs was 5.9 months (95% CI, 3.8-8.05 months). Univariate analysis identified TP53 and BRAF mutations, NF2 deletions, and BIRC2 amplifications as poor prognostic factors for PFS (P < 0.05). Conclusions This study determined the integrated genomic profiles of Asian patients with metastatic melanoma receiving CPIs and identified candidate biomarkers that reflected treatment outcomes. The molecular characterization of Asian melanoma patients receiving check-point inhibitors (CPIs) using NGS has not been reported. NRAS and TP53 mutations and NF2 deletions were significantly associated with resistance to CPIs. MYC and RPS6KB1 amplifications were associated with responsiveness to CPIs. TP53 and BRAF mutations, NF2 deletions, and BIRC2 amplifications were poor prognostic factors for progression-free survival. This is the largest integrated genomic study to date that identifying novel biomarkers of CPIs in Asian melanoma patients.
Collapse
Affiliation(s)
- S Byeon
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - H J Cho
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Precision Medicine Research Institute, Samsung Medical Center, Seoul, Korea
| | - K-T Jang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - M Kwon
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - J Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - J Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - S T Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Gogebakan KC, Berry EG, Geller AC, Sonmez K, Leachman SA, Etzioni R. Strategizing Screening for Melanoma in an Era of Novel Treatments: A Model-Based Approach. Cancer Epidemiol Biomarkers Prev 2020; 29:2599-2607. [PMID: 32958498 DOI: 10.1158/1055-9965.epi-20-0881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/05/2020] [Accepted: 09/17/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Benefit-harm tradeoffs of melanoma screening depend on disease risk and treatment efficacy. We developed a model to project outcomes of screening for melanoma in populations with different risks under historic and novel systemic treatments. METHODS Computer simulation model of a screening program with specified impact on overall and advanced-stage incidence. Inputs included meta-analyses of treatment trials, cancer registry data, and a melanoma risk prediction study RESULTS: Assuming 50% reduction in advanced stage under screening, the model projected 59 and 38 lives saved per 100,000 men under historic and novel treatments, respectively. With 10% increase in stage I, the model projects 2.9 and 4.7 overdiagnosed cases per life saved and number needed to be screened (NNS) equal to 1695 and 2632 under historical and novel treatments. When screening was performed only for the 20% of individuals with highest predicted risk, 34 and 22 lives per 100,000 were saved under historic and novel treatments. Similar results were obtained for women, but lives saved were lower. CONCLUSIONS Melanoma early detection programs must shift a substantial fraction of cases from advanced to localized stage to be sustainable. Advances in systemic therapies for melanoma might noticeably reduce benefits of screening, but restricting screening to individuals at highest risk will likely reduce intervention efforts and harms while preserving >50% of the benefit of nontargeted screening. IMPACT Our accessible modeling framework will help to guide population melanoma screening programs in an era of novel treatments for advanced disease.
Collapse
Affiliation(s)
- Kemal Caglar Gogebakan
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Elizabeth G Berry
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Alan C Geller
- Division of Public Health Practice, Harvard School of Public Health, Boston, Massachusetts
| | - Kemal Sonmez
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Department of Statistics, University of Washington, Seattle, Washington
| |
Collapse
|
19
|
The Immune System and Pathogenesis of Melanoma and Non-melanoma Skin Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1268:211-226. [DOI: 10.1007/978-3-030-46227-7_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Tang L, Long J, Li K, Zhang X, Chen X, Peng C. A novel chalcone derivative suppresses melanoma cell growth through targeting Fyn/Stat3 pathway. Cancer Cell Int 2020; 20:256. [PMID: 32565740 PMCID: PMC7302361 DOI: 10.1186/s12935-020-01336-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
Background Fyn has been documented to have oncogenic features in multiple tumors, which might be a potential therapeutic target, however, few studies on the function role of Fyn and its specific inhibitors in melanoma. Methods We investigated the impacts of Fyn and its inhibitor Lj-1-60 on melanoma through bioinformatics analysis, western blot, cell viability, cell cycle and apoptosis and xenograft tumor model as well as immunohistochemical staining. Pull-down and in vitro kinase assay were used to demonstrate Lj-1-60 targeting Fyn. Transcriptome sequencing and RT-PCR were adopted to confirm the potential mechanisms of Lj-1-60 in melanoma. Results Our findings showed that Fyn was overexpressed in melanoma cells and knocked down of Fyn suppressed the proliferation of melanoma cells. To identify the potential inhibitors of Fyn, our in-house library including total of 111,277 chemicals was conducted to vitro screening, among those compounds, 83 inhibitors were further detected to explore the effect on melanoma cells growth and discovered a novel chalcone derivative Lj-1-60 that exhibited low cellular toxicity and high anti-tumor efficacy. Lj-1-60 directly was associated with Fyn and inhibited the Fyn kinase activity with Stat3 as substrate. What's more, Lj-1-60 suppressed the proliferation of melanoma in vitro and in vivo through inducing cell cycle arrest and apoptosis. Moreover, the activation of Stat3 had also been abrogated both in Lj-1-60 treated melanoma cells or Fyn knocked down cells. Conclusion Our study revealed a novel Fyn inhibitor that could significantly suppress melanoma growth, which is a promising potential inhibitor for melanoma treatment.
Collapse
Affiliation(s)
- Ling Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan China.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000 Hunan China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Jing Long
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000 Hunan China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Keke Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000 Hunan China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Xu Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000 Hunan China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000 Hunan China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000 Hunan China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| |
Collapse
|
21
|
Zhao JJ, Wen XZ, Ding Y, Li DD, Zhu BY, Li JJ, Weng DS, Zhang X, Zhang XS. Association between immune-related adverse events and efficacy of PD-1 inhibitors in Chinese patients with advanced melanoma. Aging (Albany NY) 2020; 12:10663-10675. [PMID: 32516130 PMCID: PMC7346077 DOI: 10.18632/aging.103285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/27/2020] [Indexed: 12/26/2022]
Abstract
Programmed cell death 1 (PD-1) checkpoint inhibitor therapy leads to immune-related adverse events (irAEs). We sought to evaluate whether the development of irAEs correlates with the treatment response in Chinese patients with advanced melanoma. In this study, we conducted a retrospective study of advanced melanoma patients who received PD-1 inhibitor therapy in China between August 2014 and March 2018. A total of 93 patients treated with PD-1 inhibitors including pembrolizumab and nivolumab were enrolled. The most frequent irAEs were pruritus, rash, vitiligo, and fatigue. The median time to onset of irAEs was 6.1 weeks. The overall response rate (ORR) and disease control rate (DCR) were higher in patients with irAEs than those without irAEs (P = 0.004 and P = 0.003, respectively), and better in patients who experienced three or more irAEs than those with none (P <0.001 and P <0.001, respectively). The ORR and DCR were significantly better in patients with grade 1 to 2 irAEs when compared with those with none (P = 0.002 and P = 0.003, respectively). In addition, the median progression-free survival and overall survival were longer in patients with irAEs than in those without irAEs (P = 0.007 and P = 0.002, respectively). In conclusion, our data demonstrated that irAEs were associated with a better clinical outcome after treatment with PD-1 inhibitor therapy in Chinese patients with advanced melanoma.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xi-Zhi Wen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ya Ding
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Dan-Dan Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Bao-Yan Zhu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jing-Jing Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - De-Sheng Weng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiao-Shi Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
22
|
Wang Q, Yang Y, Yang M, Li X, Chen K. High mutation load, immune-activated microenvironment, favorable outcome, and better immunotherapeutic efficacy in melanoma patients harboring MUC16/CA125 mutations. Aging (Albany NY) 2020; 12:10827-10843. [PMID: 32491995 PMCID: PMC7346065 DOI: 10.18632/aging.103296] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/28/2020] [Indexed: 04/18/2023]
Abstract
Immunotherapies have dramatically improved survival outcome for patients with melanoma. MUC16 encodes cancer antigen 125 (CA125), which is frequently mutated in melanoma. In this study, we correlated the MUC16 mutational status with the following: tumor mutation burden (TML), multiple immune-related signals in microenvironment, deregulated pathways, survival outcome, and immunotherapeutic efficacy. We found that patients with MUC16 mutations had significantly higher TML than those without it. Enriched pro-inflammatory CD8 T cells and M1 macrophages, enhanced interferon gamma (IFNγ) and T cell-inflamed signatures, and increased cytolytic activity were associated with MUC16 mutations. Immune-suppressive M2 macrophages were enriched in patients with wild-type MUC16. Immune checkpoints expression (e.g., PD-L1, PD-1 and CTLA-4) was also elevated in patients with MUC16 mutations. Immune response relevant circuits were among the top enriched pathways in samples with MUC16 mutations. Patients with MUC16 mutations exhibited a significantly better prognosis. For patients who received immunotherapy, the presence of MUC16 mutations was associated with a better response rate and survival outcome in male patients but not in female or overall patients. These findings provide new implications for tailoring immunotherapeutic strategies for melanoma patients.
Collapse
Affiliation(s)
- Qinghua Wang
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yichen Yang
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Meng Yang
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Xiangchun Li
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| |
Collapse
|
23
|
Hirshoren N, Yoeli R, Cohen JE, Weinberger JM, Kaplan N, Merims S, Peretz T, Lotem M. Checkpoint inhibitors: Better outcomes among advanced cutaneous head and neck melanoma patients. PLoS One 2020; 15:e0231038. [PMID: 32282861 PMCID: PMC7153888 DOI: 10.1371/journal.pone.0231038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/13/2020] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate if the treatment outcomes of checkpoint inhibitors (CPI) in patients with advanced-stage skin head and neck melanoma (HNM) differs from outcomes in patients with non-HNM. DESIGN A retrospective cohort study of patients with unresectable AJCC stage III and stage IV, who received CPI between 2010 and 2017. PARTICIPANTS Overall, 122 unresectable AJCC stage III and metastatic stage IV melanoma adult patients were treated with CPI during the study period (consecutive patients). The HNM group of patients was comparable with limbs and trunk melanoma group except different distant metastatic (M1a/b/c/d) pattern (p = 0.025). MAIN OUTCOMES Comparison of overall survival and clinical response to CPI in patients with advanced-stage skin melanoma of the head and neck with non-HNM. RESULTS We analyzed 38 patients with melanoma arising in the head and neck skin regions, 33 with melanoma of limbs and 51 with trunk melanoma. Most of the head and neck patients were men (89.5%), the average age of melanoma diagnosis was 61.4±16.7 years (range 16.4-85.6). More than a third of HNM group of patients (36.8%) were 70 years and older. Overall response rate (ORR) to CPI was 50% (CR 31.6% and PR 18.4%) in the head and neck study group of patients, compared to an ORR of 36.3% and 23.5% in melanoma of the limbs and of the trunk, respectively (p = 0.03). The median overall survival of HNM group of patients was 60.2±6.3 months, CI 95% [47.7-72.7], 63% were alive at 30 months, reaching a plateau. Whereas, the median survival time of limbs and trunk melanoma were 51.2 and 53.4 months, which did not reach significance. CONCLUSIONS AND RELEVANCE Response rate to CPI is significantly improved in patients with melanoma of the head and neck and they have a trend towards improved, long standing, overall survival.
Collapse
Affiliation(s)
- Nir Hirshoren
- Department of Otolaryngology / Head & Neck Surgery, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - Roni Yoeli
- Department of Otolaryngology / Head & Neck Surgery, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - Jonathan E Cohen
- Sharett Institute of Oncology, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
- The Faculty of Medicine, The Wohl institute for Translational Medicine, Hadassah Medical Center, Hadassah Hebrew-University Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jeffrey M Weinberger
- Department of Otolaryngology / Head & Neck Surgery, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - Nadia Kaplan
- Radiology department, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - Sharon Merims
- Sharett Institute of Oncology, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - Tamar Peretz
- Sharett Institute of Oncology, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - Michal Lotem
- Sharett Institute of Oncology, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| |
Collapse
|
24
|
Granados K, Hüser L, Federico A, Sachindra S, Wolff G, Hielscher T, Novak D, Madrigal-Gamboa V, Sun Q, Vierthaler M, Larribère L, Umansky V, Utikal J. T-type calcium channel inhibition restores sensitivity to MAPK inhibitors in de-differentiated and adaptive melanoma cells. Br J Cancer 2020; 122:1023-1036. [PMID: 32063604 PMCID: PMC7109069 DOI: 10.1038/s41416-020-0751-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/19/2019] [Accepted: 01/24/2020] [Indexed: 11/25/2022] Open
Abstract
Background Drug resistance remains as one of the major challenges in melanoma therapy. It is well known that tumour cells undergo phenotypic switching during melanoma progression, increasing melanoma plasticity and resistance to mitogen-activated protein kinase inhibitors (MAPKi). Methods We investigated the melanoma phenotype switching using a partial reprogramming model to de-differentiate murine melanoma cells and target melanoma therapy adaptation against MAPKi. Results Here, we show that partially reprogrammed cells are a less proliferative and more de-differentiated cell population, expressing a gene signature for stemness and suppressing melanocyte-specific markers. To investigate adaptation to MAPKi, cells were exposed to B-Raf Proto-Oncogene (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors. De-differentiated cells became less sensitive to MAPKi, showed increased cell viability and decreased apoptosis. Furthermore, T-type calcium channels expression increased in adaptive murine cells and in human adaptive melanoma cells. Treatment with the calcium channel blocker mibefradil induced cell death, differentiation and susceptibility to MAPKi in vitro and in vivo. Conclusion In summary, we show that partial reprogramming of melanoma cells induces de-differentiation and adaptation to MAPKi. Moreover, we postulated a calcium channel blocker such as mibefradil, as a potential candidate to restore sensitivity to MAPKi in adaptive melanoma cells.
Collapse
Affiliation(s)
- Karol Granados
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany.,Department of Biochemistry, School of Medicine, University of Costa Rica (UCR), Rodrigo Facio Campus, San Pedro Montes Oca, San Jose, 2060, Costa Rica
| | - Laura Hüser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Aniello Federico
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Sachindra Sachindra
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany.,Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gretchen Wolff
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Novak
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Verónica Madrigal-Gamboa
- Department of Biochemistry, School of Medicine, University of Costa Rica (UCR), Rodrigo Facio Campus, San Pedro Montes Oca, San Jose, 2060, Costa Rica
| | - Qian Sun
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Marlene Vierthaler
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Lionel Larribère
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany.
| |
Collapse
|
25
|
Luís R, Brito C, Pojo M. Melanoma Metabolism: Cell Survival and Resistance to Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:203-223. [PMID: 32130701 DOI: 10.1007/978-3-030-34025-4_11] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cutaneous melanoma is one of the most aggressive types of cancer, presenting the highest potential to form metastases, both locally and distally, which are associated with high death rates of melanoma patients. A high somatic mutation burden is characteristic of these tumours, with most common oncogenic mutations occurring in the BRAF, NRAS and NF1 genes. These intrinsic oncogenic pathways contribute to the metabolic switch between glycolysis and oxidative phosphorylation metabolisms of melanoma, facilitating tumour progression and resulting in a high plasticity and adaptability to unfavourable conditions. Moreover, melanoma microenvironment can influence its own metabolism and reprogram several immune cell subset functions, enabling melanoma to evade the immune system. The knowledge of the biology, molecular alterations and microenvironment of melanoma has led to the development of new targeted therapies and the improvement of patient care. In this work, we reviewed the impact of melanoma metabolism in the resistance to BRAF and MEK inhibitors and immunotherapies, emphasizing the requirement to evaluate metabolic alterations upon development of novel therapeutic approaches. Here we summarized the current understanding of the impact of metabolic processes in melanomagenesis, metastasis and microenvironment, as well as the involvement of metabolic pathways in the immune modulation and resistance to targeted and immunocheckpoint therapies.
Collapse
Affiliation(s)
- Rafael Luís
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| | - Cheila Brito
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| |
Collapse
|
26
|
Bellmann L, Cappellano G, Schachtl-Riess JF, Prokopi A, Seretis A, Ortner D, Tripp CH, Brinckerhoff CE, Mullins DW, Stoitzner P. A TLR7 agonist strengthens T and NK cell function during BRAF-targeted therapy in a preclinical melanoma model. Int J Cancer 2019; 146:1409-1420. [PMID: 31702822 PMCID: PMC7003881 DOI: 10.1002/ijc.32777] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/25/2019] [Accepted: 10/31/2019] [Indexed: 12/30/2022]
Abstract
Therapeutic success of targeted therapy with BRAF inhibitors (BRAFi) for melanoma is limited by resistance development. Observations from preclinical mouse models and recent insights into the immunological effects caused by BRAFi give promise for future development of combination therapy for human melanoma. In our study, we used the transplantable D4M melanoma mouse model with the BRAFV600E mutation and concomitant PTEN loss in order to characterize alterations in tumor‐infiltrating effector immune cells when tumors become resistant to BRAFi. We found that BRAFi‐sensitive tumors displayed a pronounced inflammatory milieu characterized by high levels of cytokines and chemokines accompanied by an infiltration of T and NK cells. The tumor‐infiltrating effector cells were activated and produced high levels of IFN‐γ, TNF‐α and granzyme B. When tumors became resistant and progressively grew, they reverted to a low immunogenic state similar to untreated tumors as reflected by low mRNA levels of proinflammatory cytokines and chemokines and fewer tumor‐infiltrating T and NK cells. Moreover, these T and NK cells were functionally impaired in comparison to their counterparts in BRAFi‐sensitive tumors. Their effector cell function could be restored by additional peritumoral treatment with the TLR7 agonist imiquimod, a clinically approved agent for nonmelanoma skin cancer. Indeed, resistance to BRAFi therapy was delayed and accompanied by high numbers of activated T and NK cells in tumors. Thus, combining BRAFi with an immune stimulating agent such as a TLR ligand could be a promising alternative approach for the treatment of melanoma. What's new? While inhibitors targeting mutant BRAF proteins can induce melanoma regression, many tumors become resistant to these agents, possibly owing to immunological effects of BRAF inhibitor therapy. Here, using a preclinical mouse model, the authors show that during the early treatment phase with BRAF inhibitors, melanomas are highly immunogenic, with infiltrating T cells and natural killer cells. When resistance develops, however, tumors regress toward low immunogenicity, similar to untreated tumors. Experiments show that in the BRAF‐sensitive phase, peritumoral injection of the TLR7 ligand imiquimod preserves immunogenicity and delays resistance, thus representing a potentially effective novel therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- Lydia Bellmann
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Cappellano
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna F Schachtl-Riess
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anastasia Prokopi
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Athanasios Seretis
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Ortner
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Constance E Brinckerhoff
- Department of Medicine and Biochemistry, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Lebanon, NH
| | - David W Mullins
- Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Lebanon, NH
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Samuel E, Moore M, Voskoboynik M, Shackleton M, Haydon A. An update on adjuvant systemic therapies in melanoma. Melanoma Manag 2019; 6:MMT28. [PMID: 31807279 PMCID: PMC6891936 DOI: 10.2217/mmt-2019-0009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
There is a global increase in the incidence of melanoma, with approximately 300,000 new cases in 2018 worldwide, according to statistics from the International Agency for Research on Cancer. With this rising incidence, it is important to optimize treatment strategies in all stages of the disease to provide better patient outcomes. The role of adjuvant therapy in patients with resected stage 3 melanoma is a rapidly evolving field. Interferon was the first agent shown to have any utility in this space, however, recent advances in both targeted therapies and immunotherapies have led to a number of practice changing adjuvant trials in resected stage 3 disease.
Collapse
Affiliation(s)
- Evangeline Samuel
- Department of Medical Oncology, Monash Health, Clayton, Melbourne 3168, Australia
| | - Maggie Moore
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| | - Mark Voskoboynik
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| | - Mark Shackleton
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| | - Andrew Haydon
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| |
Collapse
|
28
|
MEK inhibition suppresses B regulatory cells and augments anti-tumor immunity. PLoS One 2019; 14:e0224600. [PMID: 31671149 PMCID: PMC6822709 DOI: 10.1371/journal.pone.0224600] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 09/29/2019] [Indexed: 12/20/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) kinase (MEK) is an integral component of the RAS pathway and a therapeutic target in RAS-driven cancers. Although tumor responses to MEK inhibition are rarely durable, MEK inhibitors have shown substantial activity and durable tumor regressions when combined with systemic immunotherapies in preclinical models of RAS-driven tumors. MEK inhibitors have been shown to potentiate anti-tumor T cell immunity, but little is known about the effects of MEK inhibition on other immune subsets, including B cells. We show here that treatment with a MEK inhibitor reduces B regulatory cells (Bregs) in vitro, and reduces the number of Bregs in tumor draining lymph nodes in a colorectal cancer model in vivo. MEK inhibition does not impede anti-tumor humoral immunity, and B cells contribute meaningfully to anti-tumor immunity in the context of MEK inhibitor therapy. Treatment with a MEK inhibitor is associated with improved T cell infiltration and an enhanced response to anti-PD1 immunotherapy. Together these data indicate that MEK inhibition may reduce Bregs while sparing anti-tumor B cell function, resulting in enhanced anti-tumor immunity.
Collapse
|
29
|
Liu FX, Ou W, Diede SJ, Whitman ED. Real-world experience with pembrolizumab in patients with advanced melanoma: A large retrospective observational study. Medicine (Baltimore) 2019; 98:e16542. [PMID: 31348273 PMCID: PMC6709121 DOI: 10.1097/md.0000000000016542] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pembrolizumab has been approved in the United States for treating advanced melanoma for >4 years. We examined real-world pembrolizumab use and associated outcomes in US oncology clinical practices, including patients who would not be eligible for clinical trials.Flatiron Health longitudinal database was used to identify adult patients with advanced melanoma initiating ≥1 dose of pembrolizumab from September 4, 2014, through December 31, 2016, with follow-up through December 31, 2017. Patients in any clinical trial during the study period were excluded. Overall survival (OS) and time on treatment from pembrolizumab initiation were analyzed using the Kaplan-Meier (KM) method. Subgroup analyses were conducted to examine OS for several patient characteristics including Eastern Cooperative Oncology Group (ECOG) performance status >1, brain metastases, and corticosteroids before pembrolizumab initiation.Pembrolizumab was administered to 315 (59%), 152 (29%), and 65 (12%) patients as first-, second-, and third-line/later therapy. Median age at pembrolizumab initiation was 68 years (range, 18-84); most patients were male (66%) and white (94%). Of those with available data, 38% had BRAF-mutant melanoma, 21% had elevated lactate dehydrogenase (LDH) level, and 23% had ECOG >1. Overall, 18% had brain metastases, and 23% were prescribed corticosteroids <3 months before initiating pembrolizumab. Median study follow-up was 12.9 months (range, 0.03-39.6). Median OS was 21.8 months (95% confidence interval [CI] 16.8-29.1); KM 1-year and 2-year survival rates were 61% and 48%, respectively; and median time on pembrolizumab treatment was 4.9 months (95% CI 3.7-5.5). Median OS for first-line pembrolizumab was not reached, and for second-line and third-line/later was 13.9 and 12.5 months, respectively (log-rank P = .0095). Significantly better OS (all P ≤.0014, log-rank test) was evident for patients with ECOG performance status (PS) of 0 to 1 (vs >1), normal (vs elevated) LDH level, and no (vs yes) corticosteroid prescription <3 months before. No difference was recorded in OS by brain metastases (log-rank P = .22) or BRAF mutation status (log-rank P = .90).These findings support effectiveness of pembrolizumab in the real-world clinical setting and provide important insights into patient characteristics and outcomes associated with pembrolizumab therapy for a heterogeneous patient population with advanced melanoma, including patients who would not be eligible for clinical trials.
Collapse
Affiliation(s)
| | | | | | - Eric D. Whitman
- Atlantic Melanoma Center, Atlantic Health System Cancer Care, Morristown, NJ
| |
Collapse
|
30
|
Exacerbation of skin psoriasis when associating an MEK inhibitor with anti-PD1 immunotherapy for metastatic melanoma. Melanoma Res 2019; 29:447-448. [PMID: 31246728 DOI: 10.1097/cmr.0000000000000601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Yang F, Wen M, Pan D, Lin X, Mo J, Dong X, Liao S, Ma Y. IL-33/ST2 Axis Regulates Vasculogenic Mimicry via ERK1/2-MMP-2/9 Pathway in Melanoma. Dermatology 2019; 235:225-233. [PMID: 30928981 DOI: 10.1159/000498857] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/08/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Melanoma, an extremely malignant form of cancer, poses a significant health risk. Vasculogenic mimicry (VM), blood vessels formed by tumor cells instead of endothelial cells, is an important factor for the rapid progression of melanoma. Interleukin (IL)-33 is an inflammatory factor commonly found in the tumor microenvironment and plays an important role in the progression of many tumors. IL-33 acts on immune cells and tumor cells through its receptor ST2. This study hypothesized that IL-33 directly affects the progression of melanoma. OBJECTIVES This study was designed to investigate the effect of IL-33 on VM of melanoma and its potential mechanism of action. METHODS The expression of ST2 was evaluated in 66 cases of melanoma collected from human patients, and the differences were analyzed. In vitro experiments were conducted to study the effects of the IL-33/ST2 axis on cell migration and invasion and to elucidate possible mechanisms. RESULTS ST2 expression is associated with that of matrix metalloproteinase (MMP)-2 and VM in melanoma of patients. IL-33 increases the abilities of proliferation, migration and invasion of melanoma cells and VM tube formation through ST2. IL-33 induces the production of MMP-2/9 via ERK1/2 phosphorylation. CONCLUSION IL-33 can directly act on melanoma cells and promote its development.
Collapse
Affiliation(s)
- Fuhan Yang
- Department of Operative Surgery, Tianjin Medical University, Tianjin, China
| | - Mingming Wen
- School of Nursing of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dayu Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xian Lin
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Jing Mo
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Shihan Liao
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yuemei Ma
- Department of Operative Surgery, Tianjin Medical University, Tianjin, China,
| |
Collapse
|
32
|
Lelliott EJ, Cullinane C, Martin CA, Walker R, Ramsbottom KM, Souza-Fonseca-Guimaraes F, Abuhammad S, Michie J, Kirby L, Young RJ, Slater A, Lau P, Meeth K, Oliaro J, Haynes N, McArthur GA, Sheppard KE. A novel immunogenic mouse model of melanoma for the preclinical assessment of combination targeted and immune-based therapy. Sci Rep 2019; 9:1225. [PMID: 30718660 PMCID: PMC6361951 DOI: 10.1038/s41598-018-37883-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Both targeted therapy and immunotherapy have been used successfully to treat melanoma, but the development of resistance and poor response rates to the individual therapies has limited their success. Designing rational combinations of targeted therapy and immunotherapy may overcome these obstacles, but requires assessment in preclinical models with the capacity to respond to both therapeutic classes. Herein, we describe the development and characterization of a novel, immunogenic variant of the BrafV600ECdkn2a−/−Pten−/− YUMM1.1 tumor model that expresses the immunogen, ovalbumin (YOVAL1.1). We demonstrate that, unlike parental tumors, YOVAL1.1 tumors are immunogenic in vivo and can be controlled by immunotherapy. Importantly, YOVAL1.1 tumors are sensitive to targeted inhibitors of BRAFV600E and MEK, responding in a manner consistent with human BRAFV600E melanoma. The YOVAL1.1 melanoma model is transplantable, immunogenic and sensitive to clinical therapies, making it a valuable platform to guide strategic development of combined targeted therapy and immunotherapy approaches in BRAFV600E melanoma.
Collapse
Affiliation(s)
- Emily J Lelliott
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Carleen Cullinane
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Claire A Martin
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rachael Walker
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Kelly M Ramsbottom
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,Division of Molecular Immunology, The Walter Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Shatha Abuhammad
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jessica Michie
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Laura Kirby
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Richard J Young
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Alison Slater
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Peter Lau
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Katrina Meeth
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jane Oliaro
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Nicole Haynes
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Department of Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Grant A McArthur
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Department of Medicine, St Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Karen E Sheppard
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia. .,Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
33
|
Comparative efficacy and safety of combination therapies for advanced melanoma: a network meta-analysis. BMC Cancer 2019; 19:43. [PMID: 30626368 PMCID: PMC6327485 DOI: 10.1186/s12885-018-5259-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/27/2018] [Indexed: 02/01/2023] Open
Abstract
Background Currently, the major treatment modalities of advanced melanoma are immune check point and mitogen-activated protein kinase (MAPK) pathway inhibitors. As lacking head-to-head randomizedcontrolled trials (RCTs) comparing immune check point and MAPK pathway inhibitors, we evaluated the efficacy and toxicity with different treatment combinations of immune check point or MAPK pathway inhibitors for advanced melanoma by network meta-analysis. Methods We searched for RCTs in Pubmed, Embase, Ovid MEDLINE, Web of Science and Cochrane Central Register for Controlled Trials through March 2017. Two reviewers performed a network meta-analysis by assessing the hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS), as well as by evaluating serious adverse events (SAEs). Results Twenty-four eligible RCTs involving 10,951 patients assigned to 11 treatment modalities were included. The combination of BRAF and MEK inhibitors demonstrated an improved OS benefit compared with all the other treatments except programmed death-1/ligand-1 (PD-1/L1) blockade because the difference in OS between the BRAF-MEK inhibitor combination and PD-1 blockade (HR: 0.85; 95% credible interval (CrI): 0.59, 1.21) was not significant. For PFS, the BRAF and MEK inhibitor combination showed a significant advantage compared with other treatments apart from the combination of PD-1/L1 and cytotoxic T lymphocyte-associated antigen-4(CTLA-4) blockade (HR:0.61; 95% CrI: 0.30, 1.25). The MEK inhibitor combined with chemotherapy was associated with the highest risk of SAEs (HR: 1.76 95% CrI: 1.21, 2.48). Conclusions The combination of BRAF and MEK inhibitors exhibited a survival advantage in OS and PFS and comparable risk of toxicity compared with chemotherapy. Electronic supplementary material The online version of this article (10.1186/s12885-018-5259-8) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
De Santis F, Del Vecchio M, Castagnoli L, De Braud F, Di Cosimo S, Franceschini D, Fucà G, Hiscott J, Malmberg KJ, McGranahan N, Pietrantonio F, Rivoltini L, Sangaletti S, Tagliabue E, Tripodo C, Vernieri C, Zitvogel L, Pupa SM, Di Nicola M. Innovative therapy, monoclonal antibodies, and beyond: Highlights from the eighth annual meeting. Cytokine Growth Factor Rev 2018; 44:1-10. [PMID: 30393044 DOI: 10.1016/j.cytogfr.2018.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The eighth annual conference of "Innovative therapy, monoclonal antibodies, and beyond" was held in Milan on Jan. 26, 2018, and hosted by Fondazione IRCCS-Istituto Nazionale dei Tumori (Fondazione IRCCS INT). The conference was divided into two main scientific sessions, of i) pre-clinical assays and novel biotargets, and ii) clinical translation, as well as a third session of presentations from young investigators, which focused on recent achievements within Fondazione IRCCS INT on immunotherapy and targeted therapies. Presentations in the first session addressed the issue of cancer immunotherapy activity with respect to tumor heterogeneity, with key topics addressing: 1) tumor heterogeneity and targeted therapy, with the definition of the evolutionary Index as an indicator of tumor heterogeneity in both space and time; 2) the analysis of cancer evolution, with the introduction of the TRACERx Consortium-a multi-million pound UK research project focused on non-small cell lung cancer (NSCLC); 3) the use of anti-estrogen agents to boost immune recognition of breast cancer cells; and 4) the high degree of functional plasticity within the NK cell repertoire, including the expansion of adaptive NK cells following viral challenges. The second session addressed: 1) the effectiveness of radiotherapy to enhance the proportion of patients responsive to immune-checkpoint blockers (ICBs); 2) the use of MDSC scores in selecting melanoma patients with high probability to be responsive to ICBs; and 3) the relevance of the gut microbiome as a predictive factor, and the potential of its perturbation in increasing the immune response rate to ICBs. Overall, a picture emerged of tumor heterogeneity as the main limitation that impairs the effectiveness of anti-cancer therapies. Thus, the choice of a specific therapy based on reproducible and selective predictive biomarkers is an urgent unmet clinical need that should be addressed in order to increase the proportion of long-term responding patients and to improve the sustainability of novel drugs.
Collapse
Affiliation(s)
- F De Santis
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Del Vecchio
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Unit of Melanoma Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - L Castagnoli
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F De Braud
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Di Cosimo
- Department of Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - D Franceschini
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Via Manzoni 56 20089 Rozzano (Milano) Italy
| | - G Fucà
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - J Hiscott
- Laboratorio Pasteur, Istituto Pasteur-Fondazione Cenci-Bolognetti, 00161 Rome, Italy
| | - K J Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Department. of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden; Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; The KG Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - N McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - F Pietrantonio
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - L Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Sangaletti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - E Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - C Tripodo
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - C Vernieri
- Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - L Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé Et de la Recherche Medicale (INSERM), Villejuif, France; Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - S M Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Di Nicola
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
35
|
Whitman ED, Liu FX, Cao X, Diede SJ, Haiderali A, Abernethy AP. Treatment patterns and outcomes for patients with advanced melanoma in US oncology clinical practices. Future Oncol 2018; 15:459-471. [PMID: 30251550 DOI: 10.2217/fon-2018-0620] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM To describe recent evolution in treatment patterns and outcomes for advanced melanoma (AMel). METHODS This retrospective observational study analyzed de-identified electronic health record data from the Flatiron Health database for 1140 adult patients who initiated first-line therapy for AMel from 1 January 2014 to 30 June 2016 with follow-up through 28 February 2017. RESULTS The most common first-line regimens were ipilimumab-based therapies (34%), anti-PD-1 monotherapy (26%) and BRAF/MEK inhibitor(s) (20%). First-line ipilimumab-based and BRAF inhibitor regimens decreased after the third quarter of 2014 (3Q2014), and by 2Q2016, 55 and 91% of BRAF-mutant and BRAF wild-type cohorts, respectively, received a first-line anti-PD-1 regimen. Median overall survival from first-line initiation for all patients was 18.8 months (95% CI: 16.3-23.3). CONCLUSION Results illustrate changing paradigms of therapy and real-world patient outcomes for AMel.
Collapse
Affiliation(s)
- Eric D Whitman
- Atlantic Melanoma Center, Atlantic Health System Cancer Care, 100 Madison Ave., Morristown, NJ 07960, USA
| | | | - Xiting Cao
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Scott J Diede
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Amin Haiderali
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Amy P Abernethy
- Flatiron Health, Inc., 233 Spring St, New York, NY 10013, USA
| |
Collapse
|
36
|
Efficacy of PD-1/PD-L1 inhibitors against pretreated advanced cancer: a systematic review and meta-analysis. Oncotarget 2018; 9:11846-11857. [PMID: 29545941 PMCID: PMC5837741 DOI: 10.18632/oncotarget.24163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/04/2017] [Indexed: 12/26/2022] Open
Abstract
Background Programmed cell death 1 (PD-1) and programmed cell death-ligand 1(PD-L1) inhibitors have captured our attention as new therapeutic options for several tumor types. Nonetheless, the differences in efficacy between PD-1/PD-L1 inhibitors and conventional treatments (chemotherapy or targeted therapy) in pretreated advanced cancer patients remain unclear. Materials and Methods A systematic literature search was conducted to identify phase III randomized controlled trials (RCTs)-based investigations of PD-1(nivolumab, pembrolizumab)/PD-L1 inhibitors (atezolizumab) against pretreated advanced cancer. We evaluated these trials for inclusion, assessed each study’s risk of bias and selected relevant data for analysis. Results The eligibility criteria were met by 5,093 patients from 8 phase III RCTs. PD-1/PD-L1 inhibitors significantly extended overall survival relative to the conventional treatment, expressed as hazard ratio [HR] (0.72, 95% CI, 0.66 to 0.77, P < 0.001) and median month difference (2.83 months, 95% CI, 1.87 to 3.78, P < 0.001). The progression-free survival HRs favored PD-1/PD-L1 inhibitors over conventional treatment (0.88; 95% CI, 0.82 to 0.95, P = 0.002), whereas median month difference was just the opposite (−0.69 months, 95% CI, −1.14 to −0.24, P < 0.001). Conclusions Among selected patients with pretreated advanced cancer, PD-1/PD-L1 inhibitors, compared with conventional treatments (chemotherapy or targeted therapy), were associated with improvement in overall survival (2.83 months) but not progression-free survival. These findings will be important in considering PD-1/PD-L1 inhibitors in the treatment of pretreated advanced cancer and have implications for future study design.
Collapse
|
37
|
Poenitzsch Strong AM, Berry SM, Beebe DJ, Li JL, Spiegelman VS. miFAST: A novel and rapid microRNA target capture method. Mol Carcinog 2018; 57:559-566. [PMID: 29350431 DOI: 10.1002/mc.22780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRNAs), small 22-25 nucleotide non-coding RNAs, play important roles in cellular and tumor biology. However, characterizing miRNA function remains challenging due to an abundance of predicted targets and an experimental bottleneck in identifying biologically relevant direct targets. Here, we developed a novel technique (miFAST) to identify direct miRNA target genes. Using miFAST, we confirmed several previously reported miR-340 target genes and identified five additional novel direct miR-340 targets in melanoma cells. This methodology can also be efficiently applied for the global characterization of miRNA targets. Utilizing miFAST to characterize direct miRNA targetomes will further our understanding of miRNA biology and function.
Collapse
Affiliation(s)
| | - Scott M Berry
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jian-Liang Li
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida
| | - Vladimir S Spiegelman
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
38
|
Audibert C, Stuntz M, Glass D. Treatment Sequencing in Advanced BRAF-Mutant Melanoma Patients: Current Practice in the United States. J Pharm Technol 2018; 34:17-23. [PMID: 34860980 DOI: 10.1177/8755122517747089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background: Treatment of advanced BRAF-mutant melanoma has changed dramatically in the past 3 years thanks to the approval of new immunotherapy and targeted therapy agents. Objectives: The goal of our survey was to investigate when immunotherapy and targeted therapy are used in the management of advanced melanoma patients and whether differences exist between the types of setting. Methods: Oncologists from academic centers, community-based centers, and private clinics were invited to participate in an online survey. Survey questions addressed the proportion of BRAF-mutant patients per treatment line, proportion of patients on targeted therapy and immunotherapy available in the United States, and reasons for prescribing each drug class. Results: A total of 101 physicians completed the survey, of which 47 worked in a private clinic, 33 in an academic center, and 21 in a community-based center. Academic center participants tended to see more severe patients (P < .001) and had more patients in second-line treatment than participants from other setting types. In addition, academic center physicians had more patients in clinical trials (P < .001), and they prescribed the ipilimumab and nivolumab combination more frequently. In terms of sequencing, all participants used targeted therapy for severe or rapidly progressing patients and immunotherapy for those who were less severe or slowly progressing. Conclusions: The findings illustrate the differences in treatment approach per type of setting, with patients in academic centers more likely to receive recently approved products or to be enrolled in clinical trials than those in community-based settings.
Collapse
|
39
|
Hernandez B, Adissu HA, Wei BR, Michael HT, Merlino G, Simpson RM. Naturally Occurring Canine Melanoma as a Predictive Comparative Oncology Model for Human Mucosal and Other Triple Wild-Type Melanomas. Int J Mol Sci 2018; 19:E394. [PMID: 29385676 PMCID: PMC5855616 DOI: 10.3390/ijms19020394] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Melanoma remains mostly an untreatable fatal disease despite advances in decoding cancer genomics and developing new therapeutic modalities. Progress in patient care would benefit from additional predictive models germane for human disease mechanisms, tumor heterogeneity, and therapeutic responses. Toward this aim, this review documents comparative aspects of human and naturally occurring canine melanomas. Clinical presentation, pathology, therapies, and genetic alterations are highlighted in the context of current basic and translational research in comparative oncology. Somewhat distinct from sun exposure-related human cutaneous melanomas, there is growing evidence that a variety of gene copy number alterations and protein structure/function mutations play roles in canine melanomas, in circumstances more analogous to human mucosal melanomas and to some extent other melanomas with murine sarcoma viral oncogene homolog B (BRAF), Neuroblastoma RAS Viral (V-Ras) Oncogene Homolog (NRAS), and neurofibromin 1 tumor suppressor NF1 triple wild-type genotype. Gaps in canine genome annotation, as well as an insufficient number and depth of sequences covered, remain considerable barriers to progress and should be collectively addressed. Preclinical approaches can be designed to include canine clinical trials addressing immune modulation as well as combined-targeted inhibition of Rat Sarcoma Superfamily/Mitogen-activated protein kinase (RAS/MAPK) and/or Phosphatidylinositol-3-Kinase/Protein Kinase B/Mammalian target of rapamycin (PI3K/AKT/mTOR) signal transduction, pathways frequently activated in both human and canine melanomas. Future investment should be aimed towards improving understanding of canine melanoma as a predictive preclinical surrogate for human melanoma and for mutually benefiting these uniquely co-dependent species.
Collapse
Affiliation(s)
- Belen Hernandez
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
- Medical Research Scholars Program, Office of Clinical Research Training and Medical Education, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Hibret A Adissu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
- Leidos Biomedical Research, Inc., Frederick, MD 21704, USA.
| | - Helen T Michael
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
- NIH Comparative Biomedical Scientist Training Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Hamid O, Puzanov I, Dummer R, Schachter J, Daud A, Schadendorf D, Blank C, Cranmer LD, Robert C, Pavlick AC, Gonzalez R, Hodi FS, Ascierto PA, Salama AKS, Margolin KA, Gangadhar TC, Wei Z, Ebbinghaus S, Ibrahim N, Ribas A. Final analysis of a randomised trial comparing pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory advanced melanoma. Eur J Cancer 2017; 86:37-45. [PMID: 28961465 DOI: 10.1016/j.ejca.2017.07.022] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/18/2017] [Indexed: 12/26/2022]
Abstract
AIM To evaluate the protocol-specified final analysis of overall survival (OS) in the KEYNOTE-002 study (NCT01704287) of pembrolizumab versus chemotherapy in patients with ipilimumab-refractory, advanced melanoma. METHODS In this randomised, phase II study, eligible patients had advanced melanoma with documented progression after two or more ipilimumab doses, previous BRAF or MEK inhibitor or both, if BRAFV600 mutant-positive. Patients were randomised to pembrolizumab 2 mg/kg or 10 mg/kg every 3 weeks or investigator-choice chemotherapy. Crossover to pembrolizumab was allowed following progression on chemotherapy. The protocol-specified final OS was performed in the intent-to-treat population. Survival was positive if p < 0.01 in one pembrolizumab arm. RESULTS A total of 180 patients were randomised to pembrolizumab 2 mg/kg, 181 to pembrolizumab 10 mg/kg and 179 to chemotherapy. At a median follow-up of 28 months (range 24.1-35.5), 368 patients died and 98 (55%) crossed over to pembrolizumab. Pembrolizumab 2 mg/kg (hazard ratio [HR] 0.86, 95% confidence interval [CI] 0.67-1.10, p = 0.117) and 10 mg/kg (0.74, 0.57-0.96, p = 0.011) resulted in a non-statistically significant improvement in OS versus chemotherapy; median OS was 13.4 (95% CI 11.0-16.4) and 14.7 (95% CI 11.3-19.5), respectively, versus 11.0 months (95% CI 8.9-13.8), with limited improvement after censoring for crossover. Two-year survival rates were 36% and 38%, versus 30%. Progression-free survival, objective response rate and duration of response improved with pembrolizumab versus chemotherapy, regardless of dose. Grade III-V treatment-related adverse events occurred in 24 (13.5%), 30 (16.8%) and 45 (26.3%) patients, respectively. CONCLUSION Improvement in OS with pembrolizumab was not statistically significant at either dose versus chemotherapy.
Collapse
Affiliation(s)
- Omid Hamid
- The Angeles Clinic and Research Institute, Los Angeles, CA, USA.
| | - Igor Puzanov
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | | - Jacob Schachter
- Ella Lemelbaum Institute of Melanoma, Sheba Medical Center, Tel Hashomer, Israel
| | - Adil Daud
- University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Lee D Cranmer
- University of Arizona Cancer Center, Tucson, AZ, USA
| | | | | | | | | | | | | | | | - Tara C Gangadhar
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ziwen Wei
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Antoni Ribas
- University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
41
|
Sengupta S. Cancer Nanomedicine: Lessons for Immuno-Oncology. Trends Cancer 2017; 3:551-560. [PMID: 28780932 DOI: 10.1016/j.trecan.2017.06.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023]
Abstract
Cancer nanotechnology and cancer immunotherapy are two parallel themes that have emerged over the last few decades in the search for a cure for cancer. Exciting applications can emerge at the intersection of these two fields. However, it is important to learn from the past successes and failures of cancer nanomedicines for its future applications in cancer immunotherapy. This review discusses the two key parameters that defined clinical success in the case of cancer nanomedicines: (i) physicochemical design principles, and (ii) clinical trial design, which are frequently overlooked in most analyses of the state of the field. Learning from the design principles that defined success for the clinically-used cancer nanomedicines can enable the design of next-generation nanomedicines that can address some of the emerging challenges in cancer immunotherapy, for example (i) enabling combinations of molecularly targeted therapies with immunotherapies that are pharmacologically incompatible; (ii) early monitoring of efficacy of immunotherapies; and (iii) personalizing an immune response to a patient's tumor. Currently, only a subset of patients treated with immunotherapy exhibit durable response; the integration of nanomedicine and immunotherapy to address the above challenges can lead to new paradigms in the treatment of cancer.
Collapse
Affiliation(s)
- Shiladitya Sengupta
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA; Dana Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
42
|
Larkin J, Minor D, D'Angelo S, Neyns B, Smylie M, Miller WH, Gutzmer R, Linette G, Chmielowski B, Lao CD, Lorigan P, Grossmann K, Hassel JC, Sznol M, Daud A, Sosman J, Khushalani N, Schadendorf D, Hoeller C, Walker D, Kong G, Horak C, Weber J. Overall Survival in Patients With Advanced Melanoma Who Received Nivolumab Versus Investigator's Choice Chemotherapy in CheckMate 037: A Randomized, Controlled, Open-Label Phase III Trial. J Clin Oncol 2017; 36:383-390. [PMID: 28671856 DOI: 10.1200/jco.2016.71.8023] [Citation(s) in RCA: 374] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Until recently, limited options existed for patients with advanced melanoma who experienced disease progression while receiving treatment with ipilimumab. Here, we report the coprimary overall survival (OS) end point of CheckMate 037, which has previously shown that nivolumab resulted in more patients achieving an objective response compared with chemotherapy regimens in ipilimumab-refractory patients with advanced melanoma. Patients and Methods Patients were stratified by programmed death-ligand 1 expression, BRAF status, and best prior cytotoxic T-lymphocyte antigen-4 therapy response, then randomly assigned 2:1 to nivolumab 3 mg/kg intravenously every 2 weeks or investigator's choice chemotherapy (ICC; dacarbazine 1,000 mg/m2 every 3 weeks or carboplatin area under the curve 6 plus paclitaxel 175 mg/m2 every 3 weeks). Patients were treated until they experienced progression or unacceptable toxicity, with follow-up of approximately 2 years. Results Two hundred seventy-two patients were randomly assigned to nivolumab (99% treated) and 133 to ICC (77% treated). More nivolumab-treated patients had brain metastases (20% v 14%) and increased lactate dehydrogenase levels (52% v 38%) at baseline; 41% of patients treated with ICC versus 11% of patients treated with nivolumab received anti-programmed death 1 agents after randomly assigned therapy. Median OS was 16 months for nivolumab versus 14 months for ICC (hazard ratio, 0.95; 95.54% CI, 0.73 to 1.24); median progression-free survival was 3.1 months versus 3.7 months, respectively (hazard ratio, 1.0; 95.1% CI, 0.78 to 1.436). Overall response rate (27% v 10%) and median duration of response (32 months v 13 months) were notably higher for nivolumab versus ICC. Fewer grade 3 and 4 treatment-related adverse events were observed in patients on nivolumab (14% v 34%). Conclusion Nivolumab demonstrated higher, more durable responses but no difference in survival compared with ICC. OS should be interpreted with caution as it was likely impacted by an increased dropout rate before treatment, which led to crossover therapy in the ICC group, and by an increased proportion of patients in the nivolumab group with poor prognostic factors.
Collapse
Affiliation(s)
- James Larkin
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - David Minor
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Sandra D'Angelo
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Bart Neyns
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Michael Smylie
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Wilson H Miller
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Ralf Gutzmer
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Gerald Linette
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Bartosz Chmielowski
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Christopher D Lao
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Paul Lorigan
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Kenneth Grossmann
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Jessica C Hassel
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Mario Sznol
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Adil Daud
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Jeffrey Sosman
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Nikhil Khushalani
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Dirk Schadendorf
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Christoph Hoeller
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Dana Walker
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - George Kong
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Christine Horak
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| | - Jeffrey Weber
- James Larkin, Royal Marsden NHS Foundation Trust, London; Paul Lorigan, The Christie National Health Service Foundation Trust, Manchester, United Kingdom; David Minor, California Pacific Medical Center Research Institute; Adil Daud, University of California San Francisco, San Francisco; Bartosz Chmielowski, University of California, Santa Monica, CA; Sandra D'Angelo, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; Jeffrey Weber, Perlmutter Cancer Center at New York University-Langone Medical Center, New York; Nikhil Khushalani, Roswell Park Cancer Institute, Buffalo, NY; Gerald Linette, Washington University, St. Louis, MO; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Mario Sznol, Yale Comprehensive Cancer Center, New Haven, CT; Jeffrey Sosman, Northwestern University, Chicago, IL; Dana Walker, George Kong, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ; Bart Neyns, University Hospital, Vrije Universiteit Brussel, Brussels, Belgium; Michael Smylie, Cross Cancer Institute, Edmonton, Alberta; Wilson H. Miller Jr, Jewish General Hospital and Segal Cancer Centre, McGill University, Montreal, Quebc, Canada; Ralf Gutzmer, Medizinische Hochschule Hannover, Hannover; Jessica C. Hassel, Nationale Centrum für Tumorerkrankungen Heidelberg, Heidelberg; Dirk Schadendorf, University Hospital Essen, Essen, Germany; and Christoph Hoeller, Medical University of Vienna, Wien, Austria
| |
Collapse
|
43
|
Tsai KK, Algazi AP. Are PD-1 antibodies safe for use in metastatic uveal melanoma? Melanoma Manag 2017; 4:79-82. [DOI: 10.2217/mmt-2017-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/04/2017] [Indexed: 11/21/2022] Open
Affiliation(s)
- Katy K Tsai
- Melanoma Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 1600 Divisadero Street, San Francisco, CA 94115, USA
| | - Alain P Algazi
- Melanoma Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 1600 Divisadero Street, San Francisco, CA 94115, USA
| |
Collapse
|
44
|
Pyo JS, Kang G. Immunotherapy in advanced melanoma: a network meta-analysis. Immunotherapy 2017; 9:471-479. [DOI: 10.2217/imt-2016-0143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The aim of this study was to compare the effects of various immunotherapeutic agents and chemotherapy for unresected or metastatic melanomas. Methods: We performed a network meta-analysis using a Bayesian statistical model to compare objective response rate (ORR) of various immunotherapies from 12 randomized controlled studies. Results: The estimated ORRs of immunotherapy and chemotherapy were 0.224 and 0.108, respectively. The ORRs of immunotherapy in untreated and pretreated patients were 0.279 and 0.176, respectively. In network meta-analysis, the odds ratios for ORR of nivolumab (1 mg/kg)/ipilmumab (3 mg/kg), pembrolizumab 10 mg/kg and nivolumab 3 mg/kg were 8.54, 5.39 and 4.35, respectively, compared with chemotherapy alone. Conclusion: Our data showed that various immunotherapies had higher ORRs rather than chemotherapy alone.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Department of Pathology, Eulji University Hospital, Eulji University School of Medicine, Daejeon, 35233, Republic of Korea
| | - Guhyun Kang
- Department of Pathology, Inje University Sanggye Paik Hospital, 1342 Dongil-ro, Nowon-gu, Seoul 139–707, Republic of Korea
| |
Collapse
|
45
|
Economou JS, Slamon DJ, Ribas A, Phelps ME. Cancer Research in the 21st Century. Ann Surg 2016; 264:555-65. [PMID: 27537535 DOI: 10.1097/sla.0000000000001926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Clancy T, Hovig E. Profiling networks of distinct immune-cells in tumors. BMC Bioinformatics 2016; 17:263. [PMID: 27377892 PMCID: PMC4932723 DOI: 10.1186/s12859-016-1141-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/20/2016] [Indexed: 11/16/2022] Open
Abstract
Background It is now clearly evident that cancer outcome and response to therapy is guided by diverse immune-cell activity in tumors. Presently, a key challenge is to comprehensively identify networks of distinct immune-cell signatures present in complex tissue, at higher-resolution and at various stages of differentiation, activation or function. This is particularly so for closely related immune-cells with diminutive, yet critical, differences. Results To predict networks of infiltrated distinct immune-cell phenotypes at higher resolution, we explored an integrated knowledge-based approach to select immune-cell signature genes integrating not only expression enrichment across immune-cells, but also an automatic capture of relevant immune-cell signature genes from the literature. This knowledge-based approach was integrated with resources of immune-cell specific protein networks, to define signature genes of distinct immune-cell phenotypes. We demonstrate the utility of this approach by profiling signatures of distinct immune-cells, and networks of immune-cells, from metastatic melanoma patients who had undergone chemotherapy. The resultant bioinformatics strategy complements immunohistochemistry from these tumors, and predicts both tumor-killing and immunosuppressive networks of distinct immune-cells in responders and non-responders, respectively. The approach is also shown to capture differences in the immune-cell networks of BRAF versus NRAS mutated metastatic melanomas, and the dynamic changes in resistance to targeted kinase inhibitors in MAPK signalling. Conclusions This integrative bioinformatics approach demonstrates that capturing the protein network signatures and ratios of distinct immune-cell in the tumor microenvironment maybe an important factor in predicting response to therapy. This may serve as a computational strategy to define network signatures of distinct immune-cells to guide immuno-pathological discovery. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1141-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Trevor Clancy
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway. .,Department of Cancer Immunology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Biomedical Research Group, Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.,Institute of Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
47
|
Albertini MR, Ranheim EA, Zuleger CL, Sondel PM, Hank JA, Bridges A, Newton MA, McFarland T, Collins J, Clements E, Henry MB, Neuman HB, Weber S, Whalen G, Galili U. Phase I study to evaluate toxicity and feasibility of intratumoral injection of α-gal glycolipids in patients with advanced melanoma. Cancer Immunol Immunother 2016; 65:897-907. [PMID: 27207605 DOI: 10.1007/s00262-016-1846-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/08/2016] [Indexed: 12/14/2022]
Abstract
Effective uptake of tumor cell-derived antigens by antigen-presenting cells is achieved pre-clinically by in situ labeling of tumor with α-gal glycolipids that bind the naturally occurring anti-Gal antibody. We evaluated toxicity and feasibility of intratumoral injections of α-gal glycolipids as an autologous tumor antigen-targeted immunotherapy in melanoma patients (pts). Pts with unresectable metastatic melanoma, at least one cutaneous, subcutaneous, or palpable lymph node metastasis, and serum anti-Gal titer ≥1:50 were eligible for two intratumoral α-gal glycolipid injections given 4 weeks apart (cohort I: 0.1 mg/injection; cohort II: 1.0 mg/injection; cohort III: 10 mg/injection). Monitoring included blood for clinical, autoimmune, and immunological analyses and core tumor biopsies. Treatment outcome was determined 8 weeks after the first α-gal glycolipid injection. Nine pts received two intratumoral injections of α-gal glycolipids (3 pts/cohort). Injection-site toxicity was mild, and no systemic toxicity or autoimmunity could be attributed to the therapy. Two pts had stable disease by RECIST lasting 8 and 7 months. Tumor nodule biopsies revealed minimal to no change in inflammatory infiltrate between pre- and post-treatment biopsies except for 1 pt (cohort III) with a post-treatment inflammatory infiltrate. Two and four weeks post-injection, treated nodules in 5 of 9 pts exhibited tumor cell necrosis without neutrophilic or lymphocytic inflammatory response. Non-treated tumor nodules in 2 of 4 evaluable pts also showed necrosis. Repeated intratumoral injections of α-gal glycolipids are well tolerated, and tumor necrosis was seen in some tumor nodule biopsies after tumor injection with α-gal glycolipids.
Collapse
Affiliation(s)
- Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- University of Wisconsin Clinical Sciences Center, Room K6/530, 600 Highland Avenue, Madison, WI, 53792, USA.
| | - Erik A Ranheim
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cindy L Zuleger
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul M Sondel
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacquelyn A Hank
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alan Bridges
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael A Newton
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Thomas McFarland
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Erin Clements
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Mary Beth Henry
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Heather B Neuman
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sharon Weber
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Giles Whalen
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - Uri Galili
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
48
|
Hölzel M, Tüting T. Inflammation-Induced Plasticity in Melanoma Therapy and Metastasis. Trends Immunol 2016; 37:364-374. [PMID: 27151281 DOI: 10.1016/j.it.2016.03.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 12/18/2022]
Abstract
Phenotype switching contributes to nongenomic heterogeneity in melanoma and other cancers. These dynamic and in part reversible phenotype changes impose diagnostic and therapeutic challenges. Understanding the reciprocal coevolution of melanoma and immune cell phenotypes during disease progression and in response to therapy is a prerequisite to improve current treatment strategies. Here we discuss how proinflammatory signals promote melanoma cell plasticity and govern interactions of melanoma and immune cells in the tumor microenvironment. We examine phenotypic plasticity and heterogeneity in different melanoma mouse models with respect to their utility for translational research and emphasize the interplay between melanoma cells and neutrophils as a critical driver of metastasis.
Collapse
Affiliation(s)
- Michael Hölzel
- Unit for RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, 53105 Bonn, Germany.
| | - Thomas Tüting
- Department of Dermatology, University Hospital Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
49
|
Abstract
Recent data have demonstrated improved survival with targeted and immune therapies in patients with advanced melanoma, leading to much excitement amongst the oncology community and the widespread use of these drugs in combination regimens. However, the place of these combination therapies in the treatment of advanced melanoma remains to be fully determined. In this perspectives article, we critically review the available data and outline the rationale for these combinations being adopted as the standard of care for patients with advanced melanoma in the future.
Collapse
Affiliation(s)
- Keiran S M Smalley
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA.
- The Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Zeynep Eroglu
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Vernon K Sondak
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| |
Collapse
|
50
|
Ebert PJR, Cheung J, Yang Y, McNamara E, Hong R, Moskalenko M, Gould SE, Maecker H, Irving BA, Kim JM, Belvin M, Mellman I. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity 2016; 44:609-621. [PMID: 26944201 DOI: 10.1016/j.immuni.2016.01.024] [Citation(s) in RCA: 541] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 12/11/2022]
Abstract
Targeted inhibition of mitogen-activated protein kinase (MAPK) kinase (MEK) can induce regression of tumors bearing activating mutations in the Ras pathway but rarely leads to tumor eradication. Although combining MEK inhibition with T-cell-directed immunotherapy might lead to more durable efficacy, T cell responses are themselves at least partially dependent on MEK activity. We show here that MEK inhibition did profoundly block naive CD8(+) T cell priming in tumor-bearing mice, but actually increased the number of effector-phenotype antigen-specific CD8(+) T cells within the tumor. MEK inhibition protected tumor-infiltrating CD8(+) T cells from death driven by chronic TCR stimulation while sparing cytotoxic activity. Combining MEK inhibition with anti-programmed death-ligand 1 (PD-L1) resulted in synergistic and durable tumor regression even where either agent alone was only modestly effective. Thus, despite the central importance of the MAP kinase pathway in some aspects of T cell function, MEK-targeted agents can be compatible with T-cell-dependent immunotherapy.
Collapse
Affiliation(s)
| | - Jeanne Cheung
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yagai Yang
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Erin McNamara
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Rebecca Hong
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | - Bryan A Irving
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jeong M Kim
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Marcia Belvin
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ira Mellman
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|