1
|
Wolf S, Madanchi M, Turko P, Hollmén M, Tugues S, von Atzigen J, Giovanoli P, Dummer R, Lindenblatt N, Halin C, Detmar M, Levesque M, Gousopoulos E. Anti-CTLA4 treatment reduces lymphedema risk potentially through a systemic expansion of the FOXP3 + T reg population. Nat Commun 2024; 15:10784. [PMID: 39737964 PMCID: PMC11686037 DOI: 10.1038/s41467-024-55002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Secondary lymphedema is a common sequel of oncologic surgery and presents a global health burden still lacking pharmacological treatment. The infiltration of the lymphedematous extremities with CD4+T cells influences lymphedema onset and emerges as a promising therapy target. Here, we show that the modulation of CD4+FOXP3+CD25+regulatory T (Treg) cells upon anti-CTLA4 treatment protects against lymphedema development in patients with melanoma and in a mouse lymphedema model. A retrospective evaluation of a melanoma patient registry reveals that anti-CTLA4 reduces lymphedema risk; in parallel, anti-CTLA4 reduces edema and improves lymphatic function in a mouse-tail lymphedema model. This protective effect of anti-CTLA4 correlates with a systemic expansion of Tregs, both in the animal model and in patients with melanoma. Our data thus show that anti-CTLA4 with its lymphedema-protective and anti-tumor properties is a promising candidate for more diverse application in the clinics.
Collapse
Affiliation(s)
- Stefan Wolf
- Division of Plastic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matiar Madanchi
- Division of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Patrick Turko
- Division of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Bioinformatic Department, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Maija Hollmén
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Sonia Tugues
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Julia von Atzigen
- Division of Plastic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Pietro Giovanoli
- Division of Plastic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Division of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Nicole Lindenblatt
- Division of Plastic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Mitchell Levesque
- Division of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Epameinondas Gousopoulos
- Division of Plastic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Adamik J, Butterfield LH. What’s next for cancer vaccines. Sci Transl Med 2022; 14:eabo4632. [DOI: 10.1126/scitranslmed.abo4632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cancer vaccines have been shown clinically to drive tumor-reactive cell activation, proliferation, and effector function. Unfortunately, tumor eradication by treatment with cancer vaccines has been unsuccessful in many patients. Critical steps are under way to improve vaccine efficacy and combine them with immunotherapy and standard-of-care treatments.
Collapse
Affiliation(s)
- Juraj Adamik
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, Suite D3500, 1 Letterman Drive, San Francisco, CA 94129, USA
| | - Lisa H. Butterfield
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, Suite D3500, 1 Letterman Drive, San Francisco, CA 94129, USA
| |
Collapse
|
3
|
Tang H, Li H, Sun Z. Targeting myeloid-derived suppressor cells for cancer therapy. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0806. [PMID: 34403220 PMCID: PMC8610166 DOI: 10.20892/j.issn.2095-3941.2020.0806] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/30/2021] [Indexed: 11/15/2022] Open
Abstract
The emergence and clinical application of immunotherapy is considered a promising breakthrough in cancer treatment. According to the literature, immune checkpoint blockade (ICB) has achieved positive clinical responses in different cancer types, although its clinical efficacy remains limited in some patients. The main obstacle to inducing effective antitumor immune responses with ICB is the development of an immunosuppressive tumor microenvironment. Myeloid-derived suppressor cells (MDSCs), as major immune cells that mediate tumor immunosuppression, are intimately involved in regulating the resistance of cancer patients to ICB therapy and to clinical cancer staging and prognosis. Therefore, a combined treatment strategy using MDSC inhibitors and ICB has been proposed and continually improved. This article discusses the immunosuppressive mechanism, clinical significance, and visualization methods of MDSCs. More importantly, it describes current research progress on compounds targeting MDSCs to enhance the antitumor efficacy of ICB.
Collapse
Affiliation(s)
- Hongchao Tang
- 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, China
| | - Hao Li
- 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, China
| | - Zhijun 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, China
- Department of Oral and Maxillofacial Head Neck Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
4
|
Schuitevoerder D, Vining CC, Tseng J. Adjuvant Therapy for Cutaneous Melanoma. Surg Oncol Clin N Am 2021; 29:455-465. [PMID: 32482320 DOI: 10.1016/j.soc.2020.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This article presents the current data supporting adjuvant therapy for patients with cutaneous melanoma. With the recent development of novel immunotherapy agents as well as targeted therapy, there are strong data to support the use of these therapies in patients at high risk of developing recurrent or metastatic disease.
Collapse
Affiliation(s)
- Darryl Schuitevoerder
- Department of Surgery, University of Chicago, 5841 South Maryland Avenue # MC5094, Chicago, IL 60637, USA
| | - Charles C Vining
- Department of Surgery, University of Chicago, 5841 South Maryland Avenue # MC5094, Chicago, IL 60637, USA
| | - Jennifer Tseng
- Department of Surgery, University of Chicago, 5841 South Maryland Avenue # MC5094, Chicago, IL 60637, USA.
| |
Collapse
|
5
|
Araujo B de Lima V, Hansen M, Spanggaard I, Rohrberg K, Reker Hadrup S, Lassen U, Svane IM. Immune Cell Profiling of Peripheral Blood as Signature for Response During Checkpoint Inhibition Across Cancer Types. Front Oncol 2021; 11:558248. [PMID: 33842304 PMCID: PMC8027233 DOI: 10.3389/fonc.2021.558248] [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: 05/01/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Despite encouraging results with immune checkpoint inhibition (ICI), a large fraction of cancer patients still does not achieve clinical benefit. Finding predictive markers in the complexity of the tumor microenvironment is a challenging task and often requires invasive procedures. In our study, we looked for putative variables related to treatment benefit among immune cells in peripheral blood across different tumor types treated with ICIs. For that, we included 33 patients with different solid tumors referred to our clinical unit for ICI. Peripheral blood mononuclear cells were isolated at baseline, 6 and 20 weeks after treatment start. Characterization of immune cells was carried out by multi-color flow cytometry. Response to treatment was assessed radiologically by RECIST 1.1. Clinical outcome correlated with a shift towards an effector-like T cell phenotype, PD-1 expression by CD8+T cells, low levels of myeloid-derived suppressor cells and classical monocytes. Dendritic cells seemed also to play a role in terms of survival. From these findings, we hypothesized that patients responding to ICI had already at baseline an immune profile, here called ‘favorable immune periphery’, providing a higher chance of benefitting from ICI. We elaborated an index comprising cell types mentioned above. This signature correlated positively with the likelihood of benefiting from the treatment and ultimately with longer survival. Our study illustrates that patients responding to ICI seem to have a pre-existing immune profile in peripheral blood that favors good outcome. Exploring this signature can help to identify patients likely to achieve benefit from ICI.
Collapse
Affiliation(s)
| | - Morten Hansen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Iben Spanggaard
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | | | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Ulrik Lassen
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| |
Collapse
|
6
|
Calabrese LH, Caporali R, Blank CU, Kirk AD. Modulating the wayward T cell: New horizons with immune checkpoint inhibitor treatments in autoimmunity, transplant, and cancer. J Autoimmun 2020; 115:102546. [PMID: 32980229 DOI: 10.1016/j.jaut.2020.102546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
The T-cell response is regulated by the balance between costimulatory and coinhibitory signals. Immune checkpoints are essential for efficient T-cell activation, but also for maintaining self-tolerance and protecting tissues from damage caused by the immune system, and for providing protective immunity. Modulating immune checkpoints can serve diametric goals, such that blocking a coinhibitory molecule can unleash anti-cancer immunity whereas stimulating the same molecule can reduce an over-reaction in autoimmune disease. The purpose of this review is to examine the regulation of T-cell costimulation and coinhibition, which is central to the processes underpinning autoimmunity, transplant rejection and immune evasion in cancer. We will focus on the immunomodulation agents that regulate these unwanted over- and under-reactions. The use of such agents has led to control of symptoms and slowing of progression in patients with rheumatoid arthritis, reduced rejection rates in transplant patients, and prolonged survival in patients with cancer. The management of immune checkpoint inhibitor treatment in certain challenging patient populations, including patients with pre-existing autoimmune conditions or transplant patients who develop cancer, as well as the management of immune-related adverse events in patients receiving antitumor therapy, is examined. Finally, the future of immune checkpoint inhibitors, including examples of emerging targets that are currently in development, as well as recent insights gained using new molecular techniques, is discussed. T-cell costimulation and coinhibition play vital roles in these diverse therapeutic areas. Targeting immune checkpoints continues to be a powerful avenue for the development of agents suitable for treating autoimmune diseases and cancers and for improving transplant outcomes. Enhanced collaboration between therapy area specialists to share learnings across disciplines will improve our understanding of the opposing effects of treatments for autoimmune disease/transplant rejection versus cancer on immune checkpoints, which has the potential to lead to improved patient outcomes.
Collapse
Affiliation(s)
| | - Roberto Caporali
- University of Milan, Department of Clinical Sciences and Community Health and Rheumatology Division, ASST Pini-CTO Hospital, Milan, Italy
| | | | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
7
|
Correlative studies investigating effects of PI3K inhibition on peripheral leukocytes in metastatic breast cancer: potential implications for immunotherapy. Breast Cancer Res Treat 2020; 184:357-364. [PMID: 32767201 DOI: 10.1007/s10549-020-05846-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 07/30/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE Patients with localized breast cancer have a 5-year survival rate > 99% compared to patients with metastatic breast cancer (MBC) that have a 5-year survival rate of ~ 27%. Unregulated PI3K/AKT signaling is a common characteristic of MBC, making it a desirable therapeutic target for tumors with activating mutations in this pathway. Interestingly, inhibition of the PI3K/AKT pathway can affect signaling in immune cells, which could potentially alter the immune phenotype of patients undergoing therapy with these drugs. The purpose of this study is to evaluate how PI3K inhibition affects the immune cells of MBC patients during treatment. METHODS We investigated the effects of PI3K inhibition on the immune cell populations in peripheral blood of MBC patients enrolled in 4 different clinical trials utilizing PI3K inhibitors. Peripheral blood was drawn at different points in patient treatment cycles to record immune cell fluctuations in response to therapy. RESULTS MBC patients who responded to treatment with a positive fold-change in cytotoxic T cell population, had an average duration of treatment response of 31.4 months. In contrast, MBC patients who responded to treatment with a negative fold-change in cytotoxic T-cell population, had an average duration of therapeutic response of 5 months. These data suggest that patients with a more robust, initial anti-tumor T cell response may have a longer therapeutic response compared to patients who do not have a robust, initial anti-tumor T cell response. CONCLUSIONS These results highlight the potential for PI3K inhibition to sensitize tumors to immune checkpoint inhibitors, thus providing additional therapeutic options for patients with MBC.
Collapse
|
8
|
Biomarkers, measured during therapy, for response of melanoma patients to immune checkpoint inhibitors: a systematic review. Melanoma Res 2020; 29:453-464. [PMID: 30855527 PMCID: PMC6727956 DOI: 10.1097/cmr.0000000000000589] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors (ICIs), which target CTLA-4 or PD-(L)1 molecules, have shown impressive therapeutic results. Durable responses, however, are only observed in a segment of the patient population and must be offset against severe off-target immune toxicity and high costs. This calls for biomarkers that predict response during ICI treatment. Although many candidate biomarkers exist, as yet, there has been no systematic overview of biomarkers predictive during. Here, we provide a systematic review of the current literature of ICI treatment to establish an overview of candidate predictive biomarkers during ICI treatment in melanoma patients. We performed a systematic Medline search (2000-2018, 1 January) on biomarkers for survival or response to ICI treatment in melanoma patients. We retrieved 735 publications, of which 79 were finally included in this systematic review. Blood markers were largely studied for CTLA-4 ICI, whereas tumor tissue markers were analyzed for PD-(L)1 ICI. Blood cytology and soluble factors were more frequently correlated to overall survival (OS) than response, indicating their prognostic rather than predictive nature. An increase in tumor-infiltrating CD8 + T-cells and a decrease in regulatory T-cells were correlated to response, in addition to mutational load, neoantigen load, and immune-related gene expression. Immune-related adverse events were also associated frequently with a favorable response and OS. This review shows the great variety of potential biomarkers published to date, in an attempt to better understand response to ICI therapy; it also highlights the candidate markers for future research. The most promising biomarkers for response to ICI treatment are the occurrence of immune-related adverse events (especially vitiligo), lowering of lactate dehydrogenase, and increase in activated CD8 + and decrease in regulatory T-cells.
Collapse
|
9
|
de Wolf ACMT, Herberts CA, Hoefnagel MHN. Dawn of Monitoring Regulatory T Cells in (Pre-)clinical Studies: Their Relevance Is Slowly Recognised. Front Med (Lausanne) 2020; 7:91. [PMID: 32300597 PMCID: PMC7142310 DOI: 10.3389/fmed.2020.00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Tregs) have a prominent role in the control of immune homeostasis. Pharmacological impact on their activity or balance with effector T cells could contribute to (impaired) clinical responses or adverse events. Monitoring treatment-related effects on T cell subsets may therefore be part of (pre-)clinical studies for medicinal products. However, the extent of immune monitoring performed in studies for marketing authorisation and the degree of correspondence with data available in the public domain is not known. We evaluated the presence of T cell immunomonitoring in 46 registration dossiers of monoclonal antibodies indicated for immune-related disorders and published scientific papers. We found that the depth of Treg analysis in registration dossiers was rather small. Nevertheless, data on treatment-related Treg effects are available in public academia-driven studies (post-registration) and suggest that Tregs may act as a biomarker for clinical responses. However, public data are fragmented and obtained with heterogeneity of experimental approaches from a diversity of species and tissues. To reveal the potential added value of T cell (and particular Treg) evaluation in (pre-)clinical studies, more cell-specific data should be acquired, at least for medicinal products with an immunomodulatory mechanism. Therefore, extensive analysis of T cell subset contribution to clinical responses and the relevance of treatment-induced changes in their levels is needed. Preferably, industry and academia should work together to obtain these data in a standardised manner and to enrich our knowledge about T cell activity in disease pathogenesis and therapies. This will ultimately elucidate the necessity of T cell subset monitoring in the therapeutic benefit-risk assessment.
Collapse
|
10
|
Zhuo Q, Yu B, Zhou J, Zhang J, Zhang R, Xie J, Wang Q, Zhao S. Lysates of Lactobacillus acidophilus combined with CTLA-4-blocking antibodies enhance antitumor immunity in a mouse colon cancer model. Sci Rep 2019; 9:20128. [PMID: 31882868 PMCID: PMC6934597 DOI: 10.1038/s41598-019-56661-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Previous reports have suggested that many gut microbiomes were associated with the development of colorectal cancer (CRC), and could modulate response to numerous forms of cancer therapy, including checkpoint blockade immunotherapy. Here we evaluated the protective efficacy of Lactobacillus acidophilus (L. acidophilus) cell lysates combined with an anti-CTL antigen-4 blocking antibody (CTLA-4 mAb) in syngeneic BALB/c mice CRC models induce by a single intraperitoneal injection of 10 mg/kg azoxymethane (AOM), followed by three cycles of 2% dextran sulfate sodium (DSS) in drinking water. In contrast to CTLA-4 mAb monotherapy, L. acidophilus lysates could attenuate the loss of body weight and the combined administration significantly protected mice against CRC development, which suggested that the lysates enhanced antitumor activity of CTLA-4 mAb in model mice. The enhanced efficacy was associated with the increased CD8 + T cell, increased effector memory T cells (CD44 + CD8 + CD62L+), decreased Treg (CD4 + CD25 + Foxp3+) and M2 macrophages (F4/80 + CD206+) in the tumor microenvironment. In addition, our results revealed that L. acidophilus lysates had an immunomodulatory effect through inhibition the M2 polarization and the IL-10 expressed levels of LPS-activated Raw264.7 macrophages. Finally, the 16S rRNA gene sequencing of fecal microbiota demonstrated that the combined administration significantly inhibited the abnormal increase in the relative abundance of proteobacteria and partly counterbalance CRC-induced dysbiosis in model mice. Overall, these data support promising clinical possibilities of L. acidophilus lysates with CTLA-4 mAb in cancer patients and the hypothesis that probiotics help shape the anticancer immune response.
Collapse
Affiliation(s)
- Qian Zhuo
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.,Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Bohai Yu
- Medical Laboratory Department, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518034, China
| | - Jing Zhou
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jingyun Zhang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Runling Zhang
- University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen, Guangdong, 518106, China
| | - Jingyan Xie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
11
|
Gravbrot N, Gilbert-Gard K, Mehta P, Ghotmi Y, Banerjee M, Mazis C, Sundararajan S. Therapeutic Monoclonal Antibodies Targeting Immune Checkpoints for the Treatment of Solid Tumors. Antibodies (Basel) 2019; 8:E51. [PMID: 31640266 PMCID: PMC6963985 DOI: 10.3390/antib8040051] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/23/2022] Open
Abstract
Recently, modulation of immune checkpoints has risen to prominence as a means to treat a number of solid malignancies, given the durable response seen in many patients and improved side effect profile compared to conventional chemotherapeutic agents. Several classes of immune checkpoint modulators have been developed. Here, we review current monoclonal antibodies directed against immune checkpoints that are employed in practice today. We discuss the history, mechanism, indications, and clinical data for each class of therapies. Furthermore, we review the challenges to durable tumor responses that are seen in some patients and discuss possible interventions to circumvent these barriers.
Collapse
Affiliation(s)
- Nicholas Gravbrot
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Kacy Gilbert-Gard
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Paras Mehta
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Yarah Ghotmi
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Madhulika Banerjee
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Christopher Mazis
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Srinath Sundararajan
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
- Texas Oncology, Dallas, TX 75251, USA.
| |
Collapse
|
12
|
Abstract
OPINION STATEMENT In recent years, the number of patients with malignant melanoma has continued to increase globally; surgery remains the first treatment option for patients with resectable melanoma. Adjuvant therapy for patients with stage III and IV melanoma following surgical resection has gradually been approved. After complete resection, these patients can probably derive significant benefit from adjuvant therapy. New treatments that improve the long-term survival of patients with unresectable advanced or metastatic melanoma are currently under evaluation in adjuvant therapy to increase relapse-free survival and overall survival. We here review several relevant clinical trials of radiotherapy, systemic immune therapies, molecular-targeted therapies, and neoadjuvant therapies in order to shed light on most suitable adjuvant therapy. The findings of this review include the following: The use of interferon-α2b will be restricted for patients with ulcerated primary melanoma in countries with no access to new drugs in adjuvant therapy. Ipilimumab should not be considered as the first-line therapy due to its lower efficacy and severe toxicity. The use of anti-programmed death-1 antibody would be a relevant adjuvant therapy for patients without BRAF mutation. If the BRAF mutation status is positive, the combination of dabrafenib and trametinib is a plausible option. The establishment of appropriate therapeutic planning and clinical endpoints in adjuvant therapy should affect the standard of care. The choice of optimal adjuvant therapy for individual patients is an important issue.
Collapse
Affiliation(s)
- Maiko Wada-Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
13
|
Butterfield LH, Vujanovic L, Santos PM, Maurer DM, Gambotto A, Lohr J, Li C, Waldman J, Chandran U, Lin Y, Lin H, Tawbi HA, Tarhini AA, Kirkwood JM. Multiple antigen-engineered DC vaccines with or without IFNα to promote antitumor immunity in melanoma. J Immunother Cancer 2019; 7:113. [PMID: 31014399 PMCID: PMC6480917 DOI: 10.1186/s40425-019-0552-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 02/27/2019] [Indexed: 02/08/2023] Open
Abstract
Background Cancer vaccines are designed to promote systemic antitumor immunity and tumor eradication. Cancer vaccination may be more efficacious in combination with additional interventions that may build on or amplify their effects. Methods Based on our previous clinical and in vitro studies, we designed an antigen-engineered DC vaccine trial to promote a polyclonal CD8+ and CD4+ T cell response against three shared melanoma antigens. The 35 vaccine recipients were then randomized to receive one month of high-dose IFNα or observation. Results The resulting clinical outcomes were 2 partial responses, 8 stable disease and 14 progressive disease among patients with measurable disease using RECIST 1.1, and, of 11 surgically treated patients with no evidence of disease (NED), 4 remain NED at a median follow-up of 3 years. The majority of vaccinated patients showed an increase in vaccine antigen-specific CD8+ and CD4+ T cell responses. The addition of IFNα did not appear to improve immune or clinical responses in this trial. Examination of the DC vaccine profiles showed that IL-12p70 secretion did not correlate with immune or clinical responses. In depth immune biomarker studies support the importance of circulating Treg and MDSC for development of antigen-specific T cell responses, and of circulating CD8+ and CD4+ T cell subsets in clinical responses. Conclusions DC vaccines are a safe and reliable platform for promoting antitumor immunity. This combination with one month of high dose IFNα did not improve outcomes. Immune biomarker analysis in the blood identified several predictive and prognostic biomarkers for further analysis, including MDSC. Trial registration NCT01622933. Electronic supplementary material The online version of this article (10.1186/s40425-019-0552-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,Department of Surgery, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.
| | - Lazar Vujanovic
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Patricia M Santos
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Deena M Maurer
- Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Joel Lohr
- Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Chunlei Li
- UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Tsinghua University School of Medicine, Beijing, China
| | - Jacob Waldman
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Uma Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huang Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hussein A Tawbi
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmad A Tarhini
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| |
Collapse
|
14
|
Coit DG, Thompson JA, Albertini MR, Barker C, Carson WE, Contreras C, Daniels GA, DiMaio D, Fields RC, Fleming MD, Freeman M, Galan A, Gastman B, Guild V, Johnson D, Joseph RW, Lange JR, Nath S, Olszanski AJ, Ott P, Gupta AP, Ross MI, Salama AK, Skitzki J, Sosman J, Swetter SM, Tanabe KK, Wuthrick E, McMillian NR, Engh AM. Cutaneous Melanoma, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 17:367-402. [PMID: 30959471 DOI: 10.6004/jnccn.2019.0018] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Cutaneous melanoma have been significantly revised over the past few years in response to emerging data on immune checkpoint inhibitor therapies and BRAF-targeted therapy. This article summarizes the data and rationale supporting extensive changes to the recommendations for systemic therapy as adjuvant treatment of resected disease and as treatment of unresectable or distant metastatic disease.
Collapse
Affiliation(s)
| | - John A Thompson
- 2Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | - William E Carson
- 4The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Carlo Contreras
- 5University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | - Ryan C Fields
- 8Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Martin D Fleming
- 9St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | - Brian Gastman
- 12Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | - Julie R Lange
- 16The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Patrick Ott
- 19Dana-Farber/Brigham and Women's Cancer Center
| | | | | | | | | | - Jeffrey Sosman
- 20Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | | |
Collapse
|
15
|
Pan M, Alavi M, Herrinton LJ. Association of Inflammatory Markers with Disease Progression in Patients with Metastatic Melanoma Treated with Immune Checkpoint Inhibitors. Perm J 2018; 22:17-149. [PMID: 29616914 DOI: 10.7812/tpp/17-149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION We investigated the effect of inflammatory biomarkers (neutrophil, platelet, and lymphocyte counts) on risk of progression in patients with metastatic melanoma treated with an immune checkpoint inhibitor targeting programmed cell death protein-1 (PD-1). METHODS This retrospective cohort study included 108 patients with malignant melanoma treated with an anti-PD-1 checkpoint inhibitor from August 2014 through December 2015. The outcome was disease progression noted on imaging or clinical examination. Follow-up began on the date of initiation of anti-PD-1 therapy and ended on the date of progression, disenrollment, death of causes other than malignant melanoma, or the end of the study in February 2017. RESULTS The median time from initiating therapy with an anti-PD-1 checkpoint inhibitor (nivolumab or pembrolizumab) to the end of follow-up was 118 days. After adjustment, baseline neutrophil and platelet counts were associated with progression. The hazard ratio (HR) for neutrophil counts ≥ 5501/μL vs ≤ 3900/μL was 2.3 (95% confidence interval [CI] = 1.2-4.6, p < 0.05). For platelet counts ≥ 304,000 vs ≤ 215,000/μL, the HR was 2.0 (CI = 1.0-3.9, p < 0.05). For lymphocyte counts ≥ 1716/μL vs ≤ 1120/μL, the HR was 0.5 (CI = 0.2-1.0, p = 0.05). CONCLUSION For patients with metastatic melanoma treated with nivolumab or pembrolizumab, higher neutrophil or platelet counts, or lower lymphocyte counts, are associated with higher risk of progression. For these patients, we recommend more frequent assessment for progression and closer follow-up, especially for patients with substantial comorbidities or poor physical performance.
Collapse
Affiliation(s)
- Minggui Pan
- Medical Oncologist at the Santa Clara Medical Center and an Adjunct Investigator for the Division of Research in Oakland, CA.
| | | | | |
Collapse
|
16
|
Tarhini A, Lin Y, Lin H, Rahman Z, Vallabhaneni P, Mendiratta P, Pingpank JF, Holtzman MP, Yusko EC, Rytlewski JA, Rao UNM, Ferris RL, Kirkwood JM. Neoadjuvant ipilimumab (3 mg/kg or 10 mg/kg) and high dose IFN-α2b in locally/regionally advanced melanoma: safety, efficacy and impact on T-cell repertoire. J Immunother Cancer 2018; 6:112. [PMID: 30352626 PMCID: PMC6199801 DOI: 10.1186/s40425-018-0428-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neoadjuvant immunotherapy utilizing novel combinations has the potential to transform the standard of care for locally/regionally advanced melanoma. We hypothesized that neoadjuvant ipilimumab in combination with high dose IFNα2b (HDI) is safe and associated with durable pathologic complete responses (pCR). METHODS Patients with locally/regionally advanced melanoma were randomized to ipilimumab 3 or 10 mg/kg × 4 doses bracketing definitive surgery, then every 12 weeks × 4. HDI was given concurrently. We evaluated the safety and efficacy of the combination with ipilimumab 3 or 10 mg/kg. The impact on T-cell fraction and clonality were investigated in tumor and blood. RESULTS Thirty patients (age 37-76), 15 each at 3 and 10 mg/kg, 18 male and 12 female were treated. Considering immune related adverse events (irAEs) of interest, more grade 3/4 irAEs were seen with ipilimumab 10 mg/kg versus 3 mg/kg (p = 0.042). Among 28 evaluable patients, 11 relapsed, of whom 5 died. Median follow-up for 17 patients who have not relapsed was 32 months. The radiologic preoperative response rate was 36% (95% CI, 21-54); 4 patients at ipilimumab 3 mg/kg and 6 at 10 mg/kg and 2 (at 10 mg/kg) later relapsed. The pCR was 32% (95% CI, 18-51); 5 patients at ipilimumab 3 mg/kg and 4 at 10 mg/kg and one (at 3 mg/kg) had a late relapse. In patients with pCR, T-cell fraction was significantly higher when measured in primary melanoma tumors (p = 0.033). Higher tumor T-cell clonality in primary tumor and more so following neoadjuvant therapy was significantly associated with improved relapse free survival. CONCLUSIONS Neoadjuvant ipilimumab-HDI was relatively safe and exhibited promising tumor response rates with an associated measurable impact on T-cell fraction and clonality. Most pCRs were durable supporting the value of pCR as a primary endpoint in neoadjuvant immunotherapy trials. TRIAL REGISTRATION ClinicalTrials.gov, NCT01608594 . Registered 31 May 2012.
Collapse
Affiliation(s)
- Ahmad Tarhini
- UPMC Hillman Cancer Center, Pittsburgh, USA. .,Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute and Case Comprehensive Cancer Center, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA.
| | - Yan Lin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | - Huang Lin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | | | | | - Prateek Mendiratta
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute and Case Comprehensive Cancer Center, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Sharma A, Subudhi SK, Blando J, Scutti J, Vence L, Wargo J, Allison JP, Ribas A, Sharma P. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3 + Regulatory T Cells (Tregs) in Human Cancers. Clin Cancer Res 2018; 25:1233-1238. [PMID: 30054281 DOI: 10.1158/1078-0432.ccr-18-0762] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/07/2018] [Accepted: 07/23/2018] [Indexed: 01/05/2023]
Abstract
PURPOSE CTLA-4 was the first inhibitory immune checkpoint to be identified. Two mAbs, ipilimumab (IgG1) and tremelimumab (IgG2), which block the function of CTLA-4, have demonstrated durable clinical activity in a subset of patients with advanced solid malignancies by augmenting effector T-cell-mediated immune responses. Studies in mice suggest that anti-CTLA-4 mAbs may also selectively deplete intratumoral FOXP3+ regulatory T cells via an Fc-dependent mechanism. However, it is unclear whether the depletion of FOXP3+ cells occurs in patients with cancer treated with anti-CTLA-4 therapies. EXPERIMENTAL DESIGN Quantitative IHC was used to evaluate the densities of intratumoral CD4+, CD8+, and FOXP3+ cells in stage-matched melanoma (n = 19), prostate cancer (n = 17), and bladder cancer (n = 9) samples treated with ipilimumab and in paired melanoma tumors (n = 18) treated with tremelimumab. These findings were corroborated with multiparametric mass cytometry analysis of tumor-infiltrating cells from paired fresh melanoma tumors (n = 5) treated with ipilimumab. RESULTS Both ipilimumab and tremelimumab increase infiltration of intratumoral CD4+ and CD8+ cells without significantly changing or depleting FOXP3+ cells within the tumor microenvironment. CONCLUSIONS Anti-CTLA-4 immunotherapy does not deplete FOXP3+ cells in human tumors, which suggests that their efficacy could be enhanced by modifying the Fc portions of the mAbs to enhance Fc-mediated depletion of intratumoral regulatory T cells.See related commentary by Quezada and Peggs, p. 1130.
Collapse
Affiliation(s)
- Anu Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Scutti
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luis Vence
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Antoni Ribas
- Department of Hematology and Oncology, University of California, Los Angeles, Los Angeles, California
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
18
|
Kitano S, Nakayama T, Yamashita M. Biomarkers for Immune Checkpoint Inhibitors in Melanoma. Front Oncol 2018; 8:270. [PMID: 30073150 PMCID: PMC6058029 DOI: 10.3389/fonc.2018.00270] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/29/2018] [Indexed: 01/05/2023] Open
Abstract
Immune checkpoint inhibitors have now become a standard therapy for malignant melanoma. However, as immunotherapies are effective in only a limited number of patients, biomarker development remains one of the most important clinical challenges. Biomarkers predicting clinical benefit facilitate appropriate selection of individualized treatments for patients and maximize clinical benefits. Many biomarkers derived from tumors and peripheral blood components have recently been reported, mainly in retrospective settings. This review summarizes the recent findings of biomarker studies for predicting the clinical benefits of immunotherapies in melanoma patients. Taking into account the complex interactions between the immune system and various cancers, it would be difficult for only one biomarker to predict clinical benefits in all patients. Many efforts to discover candidate biomarkers are currently ongoing. In the future, verification, by means of a prospective study, may allow some of these candidates to be combined into a scoring system based on bioinformatics technology.
Collapse
Affiliation(s)
- Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.,Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Takayuki Nakayama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Makiko Yamashita
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Schnell A, Schmidl C, Herr W, Siska PJ. The Peripheral and Intratumoral Immune Cell Landscape in Cancer Patients: A Proxy for Tumor Biology and a Tool for Outcome Prediction. Biomedicines 2018; 6:E25. [PMID: 29495308 PMCID: PMC5874682 DOI: 10.3390/biomedicines6010025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
Functional systemic and local immunity is required for effective anti-tumor responses. In addition to an active engagement with cancer cells and tumor stroma, immune cells can be affected and are often found to be dysregulated in cancer patients. The impact of tumors on local and systemic immunity can be assessed using a variety of approaches ranging from low-dimensional analyses that are performed on large patient cohorts to multi-dimensional assays that are technically and logistically challenging and are therefore confined to a limited sample size. Many of these strategies have been established in recent years leading to exciting findings. Not only were analyses of immune cells in tumor patients able to predict the clinical course of the disease and patients' survival, numerous studies also detected changes in the immune landscape that correlated with responses to novel immunotherapies. This review will provide an overview of established and novel tools for assessing immune cells in tumor patients and will discuss exemplary studies that utilized these techniques to predict patient outcomes.
Collapse
Affiliation(s)
- Annette Schnell
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany.
| | - Christian Schmidl
- Regensburg Centre for Interventional Immunology and University Medical Center of Regensburg, 93053 Regensburg, Germany.
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany.
- Regensburg Centre for Interventional Immunology and University Medical Center of Regensburg, 93053 Regensburg, Germany.
| | - Peter J Siska
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
20
|
Jessurun CAC, Vos JAM, Limpens J, Luiten RM. Biomarkers for Response of Melanoma Patients to Immune Checkpoint Inhibitors: A Systematic Review. Front Oncol 2017; 7:233. [PMID: 29034210 PMCID: PMC5625582 DOI: 10.3389/fonc.2017.00233] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/11/2017] [Indexed: 01/08/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs), targeting CTLA-4 or PD-1 molecules, have shown impressive therapeutic results. However, only 20–40% of advanced melanoma patients have durable responses to ICI, and these positive effects must be balanced against severe off-target immune toxicity and high costs. This urges the development of predictive biomarkers for ICI response to select patients with likely clinical benefit from treatment. Although many candidate biomarkers exist, a systematic overview of biomarkers and their usefulness is lacking. Objectives Here, we systematically review the current literature of clinical data of ICI treatment to provide an overview of candidate predictive biomarkers for ICI in melanoma patients. Methods To identify studies on biomarkers for clinical response or survival to ICI therapy in melanoma patients, we performed a systematic search in OVID MEDLINE and retrieved 429 publications, of which 67 met the eligibility criteria. Results Blood and genomic biomarkers were mainly studied for CTLA-4 ICI, while tumor tissue markers were analyzed for both CTLA-4 and PD-1 ICI. Blood cytology and soluble factors correlated more frequently to overall survival (OS) than to response, indicating their prognostic rather than predictive nature. Systemic T-cell response and regulation markers correlated to response, but progression-free survival or OS were not analyzed. Tumor tissue analyses revealed response correlations with mutational load, neoantigen load, immune-related gene expression, and CD8+ T-cell infiltration at the invasive margin. The predictive value of PD-L1 varied, possibly due to the influence of T-cell infiltration on tumor PD-L1 expression. Genomic biomarker studies addressed CTLA-4 and other immune-related genes. Conclusion This review outlines all published biomarkers for ICI therapy and highlights potential candidate markers for future research. To date, PD-L1 is the best studied biomarker for PD-1 ICI response. The most promising candidate predictive biomarkers for ICI response have not yet been identified. Variations in outcome parameters, statistical power, and analyses hampered summary of the results. Further investigation of biomarkers in larger patient cohorts using standardized objectives and outcome measures is recommended.
Collapse
Affiliation(s)
- Charissa A C Jessurun
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Julien A M Vos
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jacqueline Limpens
- Medical Library, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Rosalie M Luiten
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|