1
|
Dougan M, Nguyen LH, Buchbinder EI, Lazarus HM. Sargramostim for Prophylactic Management of Gastrointestinal Immune-Related Adverse Events of Immune Checkpoint Inhibitor Therapy for Cancer. Cancers (Basel) 2024; 16:501. [PMID: 38339253 PMCID: PMC10854719 DOI: 10.3390/cancers16030501] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy improves outcomes in several cancers. Unfortunately, many patients experience grade 3-4 treatment-related adverse events, including gastrointestinal (GI) toxicities which are common. These GI immune-related adverse events (irAEs) induced by ICIs present significant clinical challenges, require prompt intervention, and result in treatment delays or discontinuations. The treatment for these potentially severe and even fatal GI irAEs which include enterocolitis, severe diarrhea, and hepatitis may interfere with the anti-cancer approach. Sargramostim (glycosylated, yeast-derived, recombinant human GM-CSF) is an agent that has been used in clinical practice for more than 30 years with a well-recognized safety profile and has been studied in many therapeutic areas. The mechanism of action of sargramostim may treat moderate-to-severe GI irAEs without impairing the anti-cancer therapy. Some early data also suggest a potential survival benefit. Through the differentiation/maturation of monocytes, macrophages, and neutrophils and induction of anti-inflammatory T cell responses, GM-CSF aids in GI homeostasis, mucosal healing, and mucosal immunity. GM-CSF knockout mice are susceptible to severe colitis which was prevented with murine GM-CSF administration. For some patients with GI mucosa and immune cell function impairment, e.g., Crohn's disease, sargramostim reduces disease severity. In a prospective, randomized study (ECOG 1608), advanced melanoma patients had a reduction in grade 3-5 GI irAEs and less frequent colonic perforation in the sargramostim plus ipilimumab arm compared to ipilimumab alone. Sargramostim continues to be studied with ICIs for the prophylactic management of irAEs while also potentially providing a survival benefit.
Collapse
Affiliation(s)
- Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; (M.D.); (E.I.B.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Long H. Nguyen
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth I. Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; (M.D.); (E.I.B.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
Vieira GDS, Kimura TDC, Scarini JF, de Lima-Souza RA, Lavareze L, Emerick C, Gonçalves MT, Damas II, Figueiredo-Maciel T, Sales de Sá R, Aquino IG, Gonçalves de Paiva JP, Fernandes PM, Gonçalves MWA, Kowalski LP, Altemani A, Fillmore GC, Mariano FV, Egal ESA. Hematopoietic colony-stimulating factors in head and neck cancers: Recent advances and therapeutic challenges. Cytokine 2024; 173:156417. [PMID: 37944421 DOI: 10.1016/j.cyto.2023.156417] [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] [Received: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Colony-stimulating factors (CSFs) are key cytokines responsible for the production, maturation, and mobilization of the granulocytic and macrophage lineages from the bone marrow, which have been gaining attention for playing pro- and/or anti-tumorigenic roles in cancer. Head and neck cancers (HNCs) represent a group of heterogeneous neoplasms with high morbidity and mortality worldwide. Treatment for HNCs is still limited even with the advancements in cancer immunotherapy. Novel treatments for patients with recurrent and metastatic HNCs are urgently needed. This article provides an in-depth review of the role of hematopoietic cytokines such as granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), and interleukin-3 (IL-3; also known as multi-CSF) in the HNCs tumor microenvironment. We have reviewed current results from clinical trials using CSFs as adjuvant therapy to treat HNCs patients, and also clinical findings reported to date on the therapeutic application of CSFs toxicities arising from chemoradiotherapy.
Collapse
Affiliation(s)
- Gustavo de Souza Vieira
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Talita de Carvalho Kimura
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - João Figueira Scarini
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Reydson Alcides de Lima-Souza
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Luccas Lavareze
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Carolina Emerick
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Mayara Trevizol Gonçalves
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Ingrid Iara Damas
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Tayná Figueiredo-Maciel
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Raisa Sales de Sá
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Iara Gonçalves Aquino
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - João Paulo Gonçalves de Paiva
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Patrícia Maria Fernandes
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Moisés Willian Aparecido Gonçalves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery, School of Medicine, University of São Paulo (USP), São Paulo, Brazil; Department of Head and Neck Surgery and Otolaryngology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Albina Altemani
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Gary Chris Fillmore
- Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah (UU), Salt Lake City, UT, United States
| | - Fernanda Viviane Mariano
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Erika Said Abu Egal
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah (UU), Salt Lake City, UT, United States.
| |
Collapse
|
3
|
Lazarus HM, Pitts K, Wang T, Lee E, Buchbinder E, Dougan M, Armstrong DG, Paine R, Ragsdale CE, Boyd T, Rock EP, Gale RP. Recombinant GM-CSF for diseases of GM-CSF insufficiency: Correcting dysfunctional mononuclear phagocyte disorders. Front Immunol 2023; 13:1069444. [PMID: 36685591 PMCID: PMC9850113 DOI: 10.3389/fimmu.2022.1069444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Endogenous granulocyte-macrophage colony-stimulating factor (GM-CSF), identified by its ability to support differentiation of hematopoietic cells into several types of myeloid cells, is now known to support maturation and maintain the metabolic capacity of mononuclear phagocytes including monocytes, macrophages, and dendritic cells. These cells sense and attack potential pathogens, present antigens to adaptive immune cells, and recruit other immune cells. Recombinant human (rhu) GM-CSF (e.g., sargramostim [glycosylated, yeast-derived rhu GM-CSF]) has immune modulating properties and can restore the normal function of mononuclear phagocytes rendered dysfunctional by deficient or insufficient endogenous GM-CSF. Methods We reviewed the emerging biologic and cellular effects of GM-CSF. Experts in clinical disease areas caused by deficient or insufficient endogenous GM-CSF examined the role of GM-CSF in mononuclear phagocyte disorders including autoimmune pulmonary alveolar proteinosis (aPAP), diverse infections (including COVID-19), wound healing, and anti-cancer immune checkpoint inhibitor therapy. Results We discuss emerging data for GM-CSF biology including the positive effects on mitochondrial function and cell metabolism, augmentation of phagocytosis and efferocytosis, and immune cell modulation. We further address how giving exogenous rhu GM-CSF may control or treat mononuclear phagocyte dysfunction disorders caused or exacerbated by GM-CSF deficiency or insufficiency. We discuss how rhu GM-CSF may augment the anti-cancer effects of immune checkpoint inhibitor immunotherapy as well as ameliorate immune-related adverse events. Discussion We identify research gaps, opportunities, and the concept that rhu GM-CSF, by supporting and restoring the metabolic capacity and function of mononuclear phagocytes, can have significant therapeutic effects. rhu GM-CSF (e.g., sargramostim) might ameliorate multiple diseases of GM-CSF deficiency or insufficiency and address a high unmet medical need.
Collapse
Affiliation(s)
- Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, United States
| | - Katherine Pitts
- Medical Affairs, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Tisha Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elinor Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elizabeth Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - David G. Armstrong
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, United States
| | | | - Timothy Boyd
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Edwin P. Rock
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Robert Peter Gale
- Hematology Centre, Department of Immunology and Inflammation, Imperial College, London, United Kingdom
| |
Collapse
|
4
|
He K, Liu X, Hoffman RD, Shi RZ, Lv GY, Gao JL. G-CSF/GM-CSF-induced hematopoietic dysregulation in the progression of solid tumors. FEBS Open Bio 2022; 12:1268-1285. [PMID: 35612789 PMCID: PMC9249339 DOI: 10.1002/2211-5463.13445] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 11/06/2022] Open
Abstract
There are two types of abnormal hematopoiesis in solid tumor occurrence and treatment: pathological hematopoiesis, and myelosuppression induced by radiotherapy and chemotherapy. In this review, we primarily focus on the abnormal pathological hematopoietic differentiation in cancer induced by tumor-released granulocyte colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF). As key factors in hematopoietic development, G-CSF/GM-CSF are well-known facilitators of myelopoiesis and mobilization of hematopoietic stem cells (HSCs). In addition, these two cytokines can also promote or inhibit tumors, dependent on tumor type. In multiple cancer types, hematopoiesis is greatly enhanced and abnormal lineage differentiation is induced by these two cytokines. Here, dysregulated hematopoiesis induced by G-CSF/GM-CSF in solid tumors and its mechanism are summarized, and the prognostic value of G-CSF/GM-CSF-associated dysregulated hematopoiesis for tumor metastasis is also briefly highlighted.
Collapse
Affiliation(s)
- Kai He
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Xi Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Robert D Hoffman
- Yo San University of Traditional Chinese Medicine, Los Angeles, CA, 90066, USA
| | - Rong-Zhen Shi
- Tangqi Branch of Traditional Chinese Medicine Hospital of Yuhang District, Hangzhou, Zhejiang, 311106, China
| | - Gui-Yuan Lv
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou, Zhejiang, 310053, China
| | - Jian-Li Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou, Zhejiang, 310053, China
| |
Collapse
|
5
|
Tarhini AA, Joshi I, Garner F. Sargramostim and immune checkpoint inhibitors: combinatorial therapeutic studies in metastatic melanoma. Immunotherapy 2021; 13:1011-1029. [PMID: 34157863 DOI: 10.2217/imt-2021-0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The use of immune checkpoint inhibitors in patients with metastatic melanoma generates clinical benefit, including improved survival. Yet disease resistance and immune-related adverse events persist as unmet needs. Sargramostim, a yeast-derived recombinant human GM-CSF, has shown clinical activity against diverse solid tumors, including metastatic melanoma. Here we review the use of sargramostim for treatment of advanced melanoma. Potential sargramostim applications in melanoma draw on the unique ability of GM-CSF to link innate and adaptive immune responses. We review preclinical and translational data describing the mechanism of action of sargramostim and synergy with immune checkpoint inhibitors to enhance efficacy and reduce treatment-related toxicity.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Cutaneous Oncology & Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ila Joshi
- Pre-Clinical & Translational Research & Development, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| | - Fiona Garner
- Immuno-Oncology Clinical Development & Translational Medicine, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| |
Collapse
|
6
|
Lin D, He H, Sun J, He X, Long W, Cui X, Sun Y, Zhao S, Zheng X, Zeng Z, Zhang K, Wang H. Co-delivery of PSMA antigen epitope and mGM-CSF with a cholera toxin-like chimeric protein suppressed prostate tumor growth via activating dendritic cells and promoting CTL responses. Vaccine 2021; 39:1609-1620. [PMID: 33612342 DOI: 10.1016/j.vaccine.2021.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/09/2021] [Accepted: 02/02/2021] [Indexed: 01/22/2023]
Abstract
Subunit vaccines derived from tumor antigens play a role in tumor therapy because of their unique advantages. However, because of the weak immunogenicity of peptides in subunit vaccines, it is difficult to trigger an effective cytotoxic T lymphocyte (CTL) response, which is critical for cancer therapy. A requirement for the activation of CTL cells by exogenous antigens is the stimulation of antigen presenting cells (APC) with the help of adjuvants and cross-presentation to T lymphocytes. Standard nonconjugated adjuvant-peptide mixtures do not ensure co-targeting of the antigen and the adjuvant to the same APC, which limits the effects of adjuvants. In this study, a fusion protein consisting of murine granulocyte-macrophage colony stimulating factor (mGM-CSF) fused with CTA2 (A2 subunit of cholera toxin) was generated and assembled with CTB-PSMA624-632 (prostate specific membrane antigen (PSMA) peptide 624-632 fused to CTB) to obtain a cholera toxin-like protein. The chimeric protein retained the biological activity of mGM-CSF and had stronger GM1 binding activity than (CTB-PSMA624-632)5. C57BL/6J mice immunized with the CT-like chimeric protein exhibited delayed tumor growth following challenge with human PSMA-EGFP-expressing RM-1 cells. Experiment results showed that the CT-like chimeric protein could induce the maturation of DC cells and improve CTL responses. Overall, these results indicate that the nasal administration of a CT-like chimeric protein vaccine results in the development of effective immunity against prostate tumor cells and might be useful for future clinical anti-tumoral applications.
Collapse
Affiliation(s)
- Danmin Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Huafeng He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Jiajie Sun
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xianying He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Wei Long
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xiping Cui
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Yunxiao Sun
- Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, PR China
| | - Suqing Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xi Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Zheng Zeng
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, PR China
| | - Kun Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China; School of Biotechnology and Health, Wuyi University, Jiangmen 529020, PR China
| | - Huaqian Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
7
|
Abstract
GM-CSF drives the differentiation of granulocytes and monocyte/macrophages from hematopoietic stem cell progenitors. It is required for differentiating monocytes into dendritic cells (DC). Although approved for recovery of granulocytes/monocytes in patients receiving chemotherapy, G-CSF is preferred. Enthusiasm for GM-CSF monotherapy as a melanoma treatment was dampened by two large randomized trials. Although GM-CSF has been injected into tumors for many years, the efficacy of this has not been tested. There is a strong rationale for GM-CSF as a vaccine adjuvant, but it appears of benefit only for strategies that directly involve DCs, such as intratumor talimogene laherparepvec and vaccines in which DCs are loaded with antigen ex vivo and injected admixed with GM-CSF.
Collapse
Affiliation(s)
- Robert O Dillman
- Chief Medical Officer, AIVITA Biomedical, Inc. Irvine, CA 92612, USA.,Clinical Professor Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
8
|
Gill A, Gosain R, Gragg H, Bycroft R, Rai SN, Pan J, Chesney JA, Miller DM. 5-(3,3-Dimethyle-1-Triazeno) Imidazole-4-Carboxamide and Interleukin-2 Adjuvant Therapy in Resected High-Risk Primary and Regionally Metastatic Melanoma. Am J Med Sci 2019; 357:43-48. [PMID: 30611319 DOI: 10.1016/j.amjms.2018.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 09/05/2018] [Accepted: 10/19/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND In the precheckpoint inhibitor era, high-dose interferon was the only approved adjuvant therapy for high-risk melanoma. In this manuscript, we analyze the recurrence-free survival, overall survival and toxicity profile of adjuvant treatment with interleukin-2 (IL-2) and 5-(3,3-dimethyle-1-triazeno) imidazole-4-carboxamide (DTIC) for resected high-risk melanoma patients. METHODS All patients with stage IIB, IIC or stage III melanoma who were treated with DTIC/IL-2 combination therapy at a single institution from 2000 to 2010 were identified from the University of Louisville Hospital medical record. Patients received 6 months of subcutaneous IL-2 (12 × 106 units days 1-4) and intravenous DTIC (750 mg/m2 day 1 of each cycle) every 28 days for 6 cycles. Individual medical records were accessed to collect the data. RESULTS Of the 112 patients treated, all underwent surgical resection and then received adjuvant treatment. A total of 58.7% of the patients were male, 42.2% female; 99% were Caucasian. A total of 79 (72.5%) of the patients were alive at the time of analysis and 57 (47.7%) patients were currently event free. A total of 69 (63.3%) patients completed all 6 months of adjuvant combination treatment with 13.8% of the patients requiring IL-2 and 21.1% of the patients requiring DTIC dose reduction. Five year overall survival was 75.57% with recurrence-free survival of 53.05%. CONCLUSIONS For several decades, there has not been an ideal adjuvant treatment for patients with resected high risk melanoma. Our retrospective analysis suggests that combination therapy with DTIC/IL-2 is beneficial and relatively well tolerated as an alternative adjuvant treatment for patients with high-risk melanoma.
Collapse
Affiliation(s)
- Amitoj Gill
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; James Graham Brown Cancer Center, Louisville, Kentucky
| | - Rahul Gosain
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; Guthrie Corning Cancer Center, Corning, New York
| | - Hana Gragg
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; Harvard University-Brigham and Women's Hospital, Center of Excellence in Translational Research, Boston, Massachusetts
| | - Ryan Bycroft
- James Graham Brown Cancer Center, Louisville, Kentucky
| | - Shesh N Rai
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; University of Louisville, School of Public Health and Information Sciences, Louisville, Kentucky
| | - Jianmin Pan
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; University of Louisville, School of Public Health and Information Sciences, Louisville, Kentucky
| | - Jason A Chesney
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; James Graham Brown Cancer Center, Louisville, Kentucky
| | - Donald M Miller
- University of Louisville, Department of Medicine, Division of Medical Oncology/Hematology, Louisville, Kentucky; James Graham Brown Cancer Center, Louisville, Kentucky.
| |
Collapse
|
9
|
Koster BD, van den Hout MFCM, Sluijter BJR, Molenkamp BG, Vuylsteke RJCLM, Baars A, van Leeuwen PAM, Scheper RJ, Petrousjka van den Tol M, van den Eertwegh AJM, de Gruijl TD. Local Adjuvant Treatment with Low-Dose CpG-B Offers Durable Protection against Disease Recurrence in Clinical Stage I-II Melanoma: Data from Two Randomized Phase II Trials. Clin Cancer Res 2018; 23:5679-5686. [PMID: 28972083 DOI: 10.1158/1078-0432.ccr-17-0944] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/12/2017] [Accepted: 07/18/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Although risk of recurrence after surgical removal of clinical stage I-II melanoma is considerable, there is no adjuvant therapy with proven efficacy. Here, we provide clinical evidence that a local conditioning regimen, aimed at immunologic arming of the tumor-draining lymph nodes, may provide durable protection against disease recurrence (median follow-up, 88.8 months).Experimental Design: In two randomized phase II trials, patients, diagnosed with stage I-II melanoma after excision of the primary tumor, received local injections at the primary tumor excision site within 7 days preceding re-excision and sentinel lymph node (SLN) biopsy of either a saline placebo (n = 22) or low-dose CpG type B (CpG-B) with (n = 9) or without (n = 21) low-dose GM-CSF.Results: CpG-B treatment was shown to be safe, to boost locoregional and systemic immunity, to be associated with lower rates of tumor-involved SLN (10% vs. 36% in controls, P = 0.04), and, at a median follow-up of 88.8 months, to profoundly improve recurrence-free survival (P = 0.008), even for patients with histologically confirmed (i.e., pathologic) stage I-II disease (P = 0.02).Conclusions: Potentially offering durable protection, local low-dose CpG-B administration in early-stage melanoma provides an adjuvant treatment option for a large group of patients currently going untreated despite being at considerable risk for disease recurrence. Once validated in a larger randomized phase III trial, this nontoxic immunopotentiating regimen may prove clinically transformative. Clin Cancer Res; 23(19); 5679-86. ©2017 AACR.
Collapse
Affiliation(s)
- Bas D Koster
- Department of Medical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mari F C M van den Hout
- Department of Pathology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Berbel J R Sluijter
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Barbara G Molenkamp
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ronald J C L M Vuylsteke
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Arnold Baars
- Department of Pathology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Paul A M van Leeuwen
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Rik J Scheper
- Department of Pathology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - M Petrousjka van den Tol
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
10
|
Napolitano S, Brancaccio G, Argenziano G, Martinelli E, Morgillo F, Ciardiello F, Troiani T. It is finally time for adjuvant therapy in melanoma. Cancer Treat Rev 2018; 69:101-111. [PMID: 29957365 DOI: 10.1016/j.ctrv.2018.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022]
Abstract
Although melanoma is amenable to early detection, there has been no decline in the mortality rate of this disease and the prognosis of patients with high-risk primary melanoma or with macroscopic nodal involvement remains poor. The best option for patients with higher-risk melanoma is to receive effective adjuvant therapy in order to reduce their chances of recurrence. Multiple systemic therapeutic agents have been tested as adjuvant therapy for melanoma with durable benefits seen only with interferon- to date. More recently ipilimumab at the high dose of 10 mg/kg has shown a significant improvement in terms of Relapse free survival and Overall survival for stage III melanoma patients but at a significant cost in terms of immune-related toxicities. More recently, novel treatment options have emerged. The results from the latest trials with immunotherapy (PD-1 inhibitors) and molecular targeted therapy (BRAF inhibitor + MEK inhibitor) have revolutionized the management of adjuvant treatment for melanoma. As the results from these trials will mature in the next years, a change in the landscape of adjuvant treatment for melanoma is expected, resulting in new challenges in treatment decisions such as optimizing patients' selection through predictive and prognostic biomarkers, and management of treatment related adverse events, in particular immune related toxicities.
Collapse
Affiliation(s)
- S Napolitano
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - G Brancaccio
- Dermatologia e Venerologia, Dipartimento di salute mentale e fisica e medicina riabilitativa, Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - G Argenziano
- Dermatologia e Venerologia, Dipartimento di salute mentale e fisica e medicina riabilitativa, Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - E Martinelli
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - F Morgillo
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - F Ciardiello
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - T Troiani
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy.
| |
Collapse
|
11
|
Si C, Xu M, Lu M, Yu Y, Yang M, Yan M, Zhou L, Yang X. In vivo antitumor activity evaluation of cancer vaccines prepared by various antigen forms in a murine hepatocellular carcinoma model. Oncol Lett 2018; 14:7391-7397. [PMID: 29344179 PMCID: PMC5755018 DOI: 10.3892/ol.2017.7169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 10/03/2017] [Indexed: 12/23/2022] Open
Abstract
Cancer cell vaccines with strong specificity and low tolerance have been revealed to be a promising option for oncology treatment. Various antigen forms, including tumor cell lysate and glutaraldehyde-fixed tumor cells, have been intensively used in cancer vaccine preparation. However, the most effective antigen form has not yet been identified. In the present study, the antitumor efficiency of vaccines prepared by these two antigen forms was systematically investigated. Murine H22 hepatocellular carcinoma cell lysate and glutaraldehyde-fixed H22 hepatocellular carcinoma cells were conjugated with Freund's adjuvant to prepare vaccines, H22-TCL and Fixed-H22-CELL, respectively. H22-TCL and Fixed-H22-CELL were administrated by subcutaneous immunization in prophylactic and therapeutic strategies. The results of the present study revealed that H22-TCL immunization induced more significant inhibition on tumor growth and metastasis compared with Fixed-H22-CELL injection. Furthermore, histopathological observation demonstrated that H22-TCL vaccine induced larger areas of continuous necrosis within tumors compared to the Fixed-H22-CELL vaccine, which was associated with the extent of tumor inhibition. More importantly, the H22-TCL vaccine injection elicited more evident antigen-specific antibody responses compared with the Fixed-H22-CELL injection. Splenocytes from H22-TCL vaccinated mice also exhibited a more significant T lymphocytes proliferation compared with that from Fixed-H22-CELL-treated mice. All the results indicated that whole tumor cell lysate may be a more effective antigen form in cancer vaccine preparation compared with glutaraldehyde-fixed tumor cells, which elicited more marked antigen specific humoral and cellular immune responses resulted with a superior antitumor efficiency. This would have important clinical signification for cancer vaccine preparation and serve a role in prompting this to other researchers.
Collapse
Affiliation(s)
- Chunfeng Si
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Maolei Xu
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Meiyu Lu
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yan Yu
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Meizi Yang
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Miaomiao Yan
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ling Zhou
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaoping Yang
- Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
12
|
Selvarajan V, Bidkar AP, Shome R, Banerjee A, Chaubey N, Ghosh SS, Sanpui P. Studying in vitro phagocytosis of apoptotic cancer cells by recombinant GMCSF-treated RAW 264.7 macrophages. Int J Biol Macromol 2017; 102:1138-1145. [DOI: 10.1016/j.ijbiomac.2017.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 01/03/2023]
|
13
|
Mesiano G, Zini R, Montagner G, Bianchi N, Manfredini R, Chillemi A, Aglietta M, Grignani G, Lampronti I, Fiorino E, Malavasi F, Sangiolo D, Gambari R, Ferrari D. Analytic and Dynamic Secretory Profile of Patient-Derived Cytokine-Induced Killer Cells. Mol Med 2017; 23:235-246. [PMID: 28805233 DOI: 10.2119/molmed.2017.00084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/01/2017] [Indexed: 12/18/2022] Open
Abstract
Adoptive immunotherapy with Cytokine Induced Killer (CIK) cells has shown antitumor activity against several kinds of cancers in preclinical models and clinical trials. CIK cells are a subset of ex vivo expanded T lymphocytes with T-NK phenotype and MHC-unrestricted antitumor activity. Literature provides scanty information on cytokines, chemokines and growth factors secreted by CIK cells. Therefore, we investigated the secretory profile of CIK cells generated from tumor patients. The secretome analysis was performed at specific time points (day 1, day 14 and day 21) of CIK cells expansion. Mature CIK cells (day 21) produce a great variety of interleukins and secreted proteins that can be divided into 3 groups based on their secretion quantity: high (IL-13, RANTES, MIP-1α and 1β), medium (IL-1Ra, IL-5, IL-8, IL-10, IL-17, IP-10, INF-γ, VEGF and GMCSF) and low (IL-1β, IL-4, IL-6, IL-7, IL-9, IL-12, IL-15, Eotaxin, PDGF-bb, FGF basic, G-CSF and MCP-1) secreted. Moreover, comparing PBMC (day 1) and mature CIK cells (day 14 and 21) secretome, we observed that IL-5, IL-10, IL-13, GM-CSF, VEGF resulted greatly up-regulated, while IL-1β, IL-6, IL-8, IL-15, IL-17, eotaxin, MCP-1, and RANTES were down-regulated. We also performed a gene expression profile analysis of patient-derived CIK cells showing that mRNA for the different cytokines and secreted proteins were modulated during PBMC to CIK differentiation. We highlighted previously unknown secretory properties and provided for the first time a comprehensive molecular characterization of CIK cells. Our findings provide rationale to explore the functional implications and possible therapeutic modulation of CIK secretome.
Collapse
Affiliation(s)
- Giulia Mesiano
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Montagner
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonella Chillemi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Massimo Aglietta
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Giovanni Grignani
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Ilaria Lampronti
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Erika Fiorino
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Fabio Malavasi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Dario Sangiolo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Roberto Gambari
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Davide Ferrari
- Department of Life Science and Biotechnology, Sections of Microbiology and Applied Pathology; Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy.,Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
14
|
Spitler LE, Cao H, Piironen T, Whiteside TL, Weber RW, Cruickshank S. Biological Effects of Anti-Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Antibody Formation in Patients Treated With GM-CSF (Sargramostim) as Adjuvant Therapy of Melanoma. Am J Clin Oncol 2017; 40:207-213. [PMID: 25286079 PMCID: PMC4385005 DOI: 10.1097/coc.0000000000000124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We investigated the development of binding and neutralizing antibodies to granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients receiving prolonged therapy with GM-CSF as adjuvant therapy of melanoma and the impact of these antibodies on biological effects. METHODS Fifty-three patients with high-risk melanoma that had been surgically excised were treated with GM-CSF, 125 μg/m daily for 14 days every 28 days for 1 year after surgical resection of disease. Serum samples for antibodies to GM-CSF were measured before treatment and on study days 155 and 351. Blood draws for testing biological effects were keyed to GM-CSF administration: days 0 (before), 15 (after 14 d on GM-CSF), 29 (after 14 d off GM-CSF), 155, and 351 (after 14 d on GM-CSF in the sixth and 13th cycle of treatment). RESULTS Of 53 patients enrolled, 43 were evaluable for the development of anti-GM-CSF antibodies. Of these, 93% developed binding antibodies and 42% developed both binding and neutralizing antibodies. The increase in the white blood cell count, percent eosinophils, or neopterin levels engendered by GM-CSF administration was abrogated or markedly decreased in patients with neutralizing antibodies but not in patients who developed only binding antibodies. CONCLUSIONS Ninety-three percent of patients with melanoma treated with GM-CSF as adjuvant therapy develop antibodies to GM-CSF. In those with neutralizing antibodies, a diminution of the biological effects of GM-CSF was observed. The development of neutralizing antibodies might also abrogate the potential clinical benefit of this treatment and should be considered in the design of future clinical trials.
Collapse
Affiliation(s)
- Lynn E. Spitler
- Northern California Melanoma Center, St. Mary’s Medical Center, San Francisco, CA
| | - Huynh Cao
- Northern California Melanoma Center, St. Mary’s Medical Center, San Francisco, CA
| | | | | | - Robert W. Weber
- Northern California Melanoma Center, St. Mary’s Medical Center, San Francisco, CA
| | | |
Collapse
|
15
|
Vanitha S, Chaubey N, Ghosh SS, Sanpui P. Recombinant human granulocyte macrophage colony stimulating factor (hGM-CSF): Possibility of nanoparticle-mediated delivery in cancer immunotherapy. Bioengineered 2016; 8:120-123. [PMID: 27459024 DOI: 10.1080/21655979.2016.1212136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Most of the cancer treatment strategies from chemotherapy to radiotherapy render cancer cells apoptotic and these apoptotic cancer cells accumulate at the tumor sites. The accumulation of apoptotic cancer cells often result in inflammation and autoimmune responses causing serious health implications. Macrophages, which are effective immune combatants, can help in the clearance of these deleterious occupants. Granulocyte macrophage colony stimulating factor (GM-CSF) is a key cytokine, modulator of immune system and responsible for growth and differentiation of granulocytes and macrophages. In this regard, supply of recombinant GM-CSF can enhance the capability of macrophages for clearance of apoptotic cancer cells. However, delivery of the cytokine in vivo can suffer from certain disadvantages like faster depletion, less stability and low targeting efficiency. We believe that the stability and sustained release of GM-CSF can be improved through its encapsulation inside appropriately designed nanoparticles.
Collapse
Affiliation(s)
- Selvarajan Vanitha
- a Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Assam , India
| | - Nidhi Chaubey
- a Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Assam , India
| | - Siddhartha S Ghosh
- a Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Assam , India.,b Centre for Nanotechnology, Indian Institute of Technology Guwahati , Assam , India
| | - Pallab Sanpui
- b Centre for Nanotechnology, Indian Institute of Technology Guwahati , Assam , India
| |
Collapse
|
16
|
Hoeller C, Michielin O, Ascierto PA, Szabo Z, Blank CU. Systematic review of the use of granulocyte-macrophage colony-stimulating factor in patients with advanced melanoma. Cancer Immunol Immunother 2016; 65:1015-34. [PMID: 27372293 PMCID: PMC4995227 DOI: 10.1007/s00262-016-1860-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/15/2016] [Indexed: 12/24/2022]
Abstract
Several immunomodulatory checkpoint inhibitors have been approved for the treatment of patients with advanced melanoma, including ipilimumab, nivolumab and pembrolizumab. Talimogene laherparepvec is the first oncolytic virus to gain regulatory approval in the USA; it is also approved in Europe. Talimogene laherparepvec expresses granulocyte–macrophage colony-stimulating factor (GM-CSF), and with other GM-CSF-expressing oncolytic viruses in development, understanding the clinical relevance of this cytokine in treating advanced melanoma is important. Results of trials of GM-CSF in melanoma have been mixed, and while GM-CSF has the potential to promote anti-tumor responses, some preclinical data suggest that GM-CSF may sometimes promote tumor growth. GM-CSF has not been approved as a melanoma treatment. We undertook a systematic literature review of studies of GM-CSF in patients with advanced melanoma (stage IIIB–IV). Of the 503 articles identified, 26 studies met the eligibility criteria. Most studies investigated the use of GM-CSF in combination with another treatment, such as peptide vaccines or chemotherapy, or as an adjuvant to surgery. Some clinical benefit was reported in patients who received GM-CSF as an adjuvant to surgery, or in combination with other treatments. In general, outcomes for patients receiving peptide vaccines were not improved with the addition of GM-CSF. GM-CSF may be a valuable therapeutic adjuvant; however, further studies are needed, particularly head-to-head comparisons, to confirm the optimal dosing regimen and clinical effectiveness in patients with advanced melanoma.
Collapse
Affiliation(s)
- Christoph Hoeller
- Department of Dermatology, Medical University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital, Champ de l'Air, Rue du Bugnon 21, 1011, Lausanne, Switzerland.,Ludwig Centre and Swiss Institute of Bioinformatics, Génopode Building, 1015, Lausanne, Switzerland
| | - Paolo A Ascierto
- Istituto Nazionale Tumori, Fondazione 'G. Pascale', Via Mariano Semmola, 52, 80131, Naples, Italy
| | - Zsolt Szabo
- Clinical Development, Amgen Europe GmbH, Dammstrasse 23, 6300, Zug, Switzerland
| | - Christian U Blank
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| |
Collapse
|
17
|
Lawson DH, Lee S, Zhao F, Tarhini AA, Margolin KA, Ernstoff MS, Atkins MB, Cohen GI, Whiteside TL, Butterfield LH, Kirkwood JM. Randomized, Placebo-Controlled, Phase III Trial of Yeast-Derived Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Versus Peptide Vaccination Versus GM-CSF Plus Peptide Vaccination Versus Placebo in Patients With No Evidence of Disease After Complete Surgical Resection of Locally Advanced and/or Stage IV Melanoma: A Trial of the Eastern Cooperative Oncology Group-American College of Radiology Imaging Network Cancer Research Group (E4697). J Clin Oncol 2015; 33:4066-76. [PMID: 26351350 PMCID: PMC4669592 DOI: 10.1200/jco.2015.62.0500] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE We conducted a double-blind, placebo-controlled trial to evaluate the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) and peptide vaccination (PV) on relapse-free survival (RFS) and overall survival (OS) in patients with resected high-risk melanoma. PATIENTS AND METHODS Patients with completely resected stage IV or high-risk stage III melanoma were grouped by human leukocyte antigen (HLA) -A2 status. HLA-A2-positive patients were randomly assigned to receive GM-CSF, PV, both, or placebo; HLA-A2-negative patients, GM-CSF or placebo. Treatment lasted for 1 year or until recurrence. Efficacy analyses were conducted in the intent-to-treat population. RESULTS A total of 815 patients were enrolled. There were no significant improvements in OS (stratified log-rank P = .528; hazard ratio, 0.94; 95% repeated CI, 0.77 to 1.15) or RFS (P = .131; hazard ratio, 0.88; 95% CI, 0.74 to 1.04) in the patients assigned to GM-CSF (n = 408) versus those assigned to placebo (n = 407). The median OS times with GM-CSF versus placebo treatments were 69.6 months (95% CI, 53.4 to 83.5 months) versus 59.3 months (95% CI, 44.4 to 77.3 months); the 5-year OS probability rates were 52.3% (95% CI, 47.3% to 57.1%) versus 49.4% (95% CI, 44.3% to 54.3%), respectively. The median RFS times with GM-CSF versus placebo were 11.4 months (95% CI, 9.4 to 14.8 months) versus 8.8 months (95% CI, 7.5 to 11.2 months); the 5-year RFS probability rates were 31.2% (95% CI, 26.7% to 35.9%) versus 27.0% (95% CI, 22.7% to 31.5%), respectively. Exploratory analyses showed a trend toward improved OS in GM-CSF-treated patients with resected visceral metastases. When survival in HLA-A2-positive patients who received PV versus placebo was compared, RFS and OS were not significantly different. Treatment-related grade 3 or greater adverse events were similar between GM-CSF and placebo groups. CONCLUSION Neither adjuvant GM-CSF nor PV significantly improved RFS or OS in patients with high-risk resected melanoma. Exploratory analyses suggest that GM-CSF may be beneficial in patients with resected visceral metastases; this observation requires prospective validation.
Collapse
Affiliation(s)
- David H Lawson
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD.
| | - Sandra Lee
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Fengmin Zhao
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Ahmad A Tarhini
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Kim A Margolin
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Marc S Ernstoff
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Michael B Atkins
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Gary I Cohen
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Theresa L Whiteside
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Lisa H Butterfield
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - John M Kirkwood
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| |
Collapse
|
18
|
Kwek SS, Kahn J, Greaney SK, Lewis J, Cha E, Zhang L, Weber RW, Leonard L, Markovic SN, Fong L, Spitler LE. GM-CSF and ipilimumab therapy in metastatic melanoma: Clinical outcomes and immunologic responses. Oncoimmunology 2015; 5:e1101204. [PMID: 27141383 DOI: 10.1080/2162402x.2015.1101204] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/22/2022] Open
Abstract
We conducted a phase II clinical trial of anti-CTLA-4 antibody (ipilimumab) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in 22 patients with metastatic melanoma and determined clinical outcomes and immunologic responses. The treatment consisted of a 3-mo induction with ipilimumab at 10 mg/kg administered every 3 weeks for four doses in combination with GM-CSF at 125 µg/m2 for 14 d beginning on the day of the ipilimumab infusion and then GM-CSF for 3 mo on the same schedule without ipilimumab. This was followed by maintenance therapy with the combination every 3 mo for up to 2 y or until disease progression or unacceptable toxicity. Blood samples for determination of immune subsets were obtained before treatment, at week 3 (end of cycle 1) and at week 6 (end of cycle 2). Blood samples were also obtained from seven subjects who were cancer-free. The immune response disease control (irDC) rate at 24 weeks was 41% and the overall response rate (ORR) was 32%. The median progression free-survival (PFS) was 3.5 mo and the median overall survival (OS) was 21.1 mo. 41% of the patients experienced Grade 3 to 4 adverse events. We conclude that this combination is safe and the results suggest the combination may be more effective than ipilimumab monotherapy. Further, the results suggest that lower levels of CD4+ effector T cells but higher levels of CD8+ T cells expressing PD-1 at pre-treatment could be a potential biomarker for disease control in patients who receive immunotherapy with ipilimumab and GM-CSF. Further trials of this combination are warranted.
Collapse
Affiliation(s)
- Serena S Kwek
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - James Kahn
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Samantha K Greaney
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Jera Lewis
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Edward Cha
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Li Zhang
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Robert W Weber
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Lonnie Leonard
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Svetomir N Markovic
- Division of Hematology, Oncology Department , Mayo Clinic , Rochester, MN, USA
| | - Lawrence Fong
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Lynn E Spitler
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| |
Collapse
|
19
|
Tabolacci C, Cordella M, Turcano L, Rossi S, Lentini A, Mariotti S, Nisini R, Sette G, Eramo A, Piredda L, De Maria R, Facchiano F, Beninati S. Aloe-emodin exerts a potent anticancer and immunomodulatory activity on BRAF-mutated human melanoma cells. Eur J Pharmacol 2015; 762:283-92. [PMID: 26048310 DOI: 10.1016/j.ejphar.2015.05.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 12/18/2022]
Abstract
Aim of this study was to extend the knowledge on the antineoplastic effect of aloe-emodin (AE), a natural hydroxyanthraquinone compound, both in metastatic human melanoma cell lines and in primary stem-like cells (melanospheres). Treatment with AE caused reduction of cell proliferation and induction of SK-MEL-28 and A375 cells differentiation, characterized by a marked increase of transamidating activity of transglutaminase whose expression remained unmodified. In vitro antimetastatic property of AE was evaluated by adhesion and Boyden chamber invasion assays. The effect of AE on melanoma cytokines/chemokines production was determined by a multiplex assay: interestingly AE showed an immunomodulatory activity through GM-CSF and IFN-γ production. We report also that AE significantly reduced the proliferation, stemness and invasive potential of melanospheres. Moreover, AE treatment significantly enhanced dabrafenib (a BRAF inhibitor) antiproliferative activity in BRAF mutant cell lines. Our results confirm that AE possesses remarkable antineoplastic properties against melanoma cells, indicating this anthraquinone as a promising agent for differentiation therapy of cancer, or as adjuvant in chemotherapy and targeted therapy. Further, its mechanisms of action support a potential efficacy of AE treatment to counteract resistance of BRAF-mutated melanoma cells to target therapy.
Collapse
Affiliation(s)
- Claudio Tabolacci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy; Department of Biology, University "Tor Vergata", Rome, Italy
| | - Martina Cordella
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lorenzo Turcano
- Department of Biology, University "Tor Vergata", Rome, Italy
| | - Stefania Rossi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Sabrina Mariotti
- Department of Infectious, Parasitic, and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Nisini
- Department of Infectious, Parasitic, and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Adriana Eramo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Piredda
- Department of Biology, University "Tor Vergata", Rome, Italy
| | | | - Francesco Facchiano
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Simone Beninati
- Department of Biology, University "Tor Vergata", Rome, Italy
| |
Collapse
|
20
|
Aliper AM, Frieden-Korovkina VP, Buzdin A, Roumiantsev SA, Zhavoronkov A. A role for G-CSF and GM-CSF in nonmyeloid cancers. Cancer Med 2014; 3:737-46. [PMID: 24692240 PMCID: PMC4303143 DOI: 10.1002/cam4.239] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 02/08/2014] [Accepted: 03/04/2014] [Indexed: 12/17/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) modulate progression of certain solid tumors. The G-CSF- or GM-CSF-secreting cancers, albeit not very common are, however, among the most rapidly advancing ones due to a cytokine-mediated immune suppression and angiogenesis. Similarly, de novo angiogenesis and vasculogenesis may complicate adjuvant use of recombinant G-CSF or GM-CSF thus possibly contributing to a cancer relapse. Rapid diagnostic tools to differentiate G-CSF- or GM-CSF-secreting cancers are not well developed therefore hindering efforts to individualize treatments for these patients. Given an increasing utilization of adjuvant G-/GM-CSF in cancer therapy, we aimed to summarize recent studies exploring their roles in pathophysiology of solid tumors and to provide insights into some complexities of their therapeutic applications.
Collapse
Affiliation(s)
- Alexander M Aliper
- Federal Clinical Research Center of Pediatric Hematology, Oncology and Immunology, Samory Mashela 1, Moscow, 117198, Russia
| | | | | | | | | |
Collapse
|
21
|
Hughes T, Coffin RS, Lilley CE, Ponce R, Kaufman HL. Critical analysis of an oncolytic herpesvirus encoding granulocyte-macrophage colony stimulating factor for the treatment of malignant melanoma. Oncolytic Virother 2014; 3:11-20. [PMID: 27512660 PMCID: PMC4918360 DOI: 10.2147/ov.s36701] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Oncolytic viruses that selectively lyse tumor cells with minimal damage to normal cells are a new area of therapeutic development in oncology. An attenuated herpesvirus encoding the granulocyte-macrophage colony stimulating factor (GM-CSF), known as talimogene laherparepvec (T-VEC), has been identified as an attractive oncolytic virus for cancer therapy based on preclinical tumor studies and results from early-phase clinical trials and a large randomized Phase III study in melanoma. In this review, we discuss the basic biology of T-VEC, describe the role of GM-CSF as an immune adjuvant, summarize the preclinical data, and report the outcomes of published clinical trials using T-VEC. The emerging data suggest that T-VEC is a safe and potentially effective antitumor therapy in malignant melanoma and represents the first oncolytic virus to demonstrate therapeutic activity against human cancer in a randomized, controlled Phase III study.
Collapse
Affiliation(s)
- Tasha Hughes
- Departments of General Surgery and Immunology and Microbiology, Rush University Medical Center, Chicago IL, USA
| | - Robert S Coffin
- BioVex, Inc, a subsidiary of Amgen, Inc, Sherman Oaks, CA, USA
| | | | - Rafael Ponce
- BioVex, Inc, a subsidiary of Amgen, Inc, Sherman Oaks, CA, USA
| | - Howard L Kaufman
- Departments of General Surgery and Immunology and Microbiology, Rush University Medical Center, Chicago IL, USA
| |
Collapse
|
22
|
Fleming NH, Tian J, Vega-Saenz de Miera E, Gold H, Darvishian F, Pavlick AC, Berman RS, Shapiro RL, Polsky D, Osman I. Impact of age on the management of primary melanoma patients. Oncology 2013; 85:173-81. [PMID: 24008821 DOI: 10.1159/000351499] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 04/18/2013] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Age is an understudied factor when considering treatment options for melanoma. Here, we examine the impact of age on primary melanoma treatment in a prospective cohort of patients. METHODS We used logistic regression models to examine the associations between age and initial treatment, using recurrence and melanoma-specific survival as endpoints. RESULTS 444 primary melanoma patients were categorized into three groups by age at diagnosis: 19-45 years (24.3%), 46-70 (50.2%), and 71-95 (25.5%). In multivariate models, older patients experienced a higher risk of recurrence (hazard ratio 3.34, 95% confidence interval, CI, 1.53-7.25; p < 0.01). No significant differences were observed in positive biopsy margin rates or extent of surgical margins across age groups. Patients in the middle age group were more likely to receive adjuvant therapy than those in the older group (odds ratio 2.78, 95% CI 1.19-6.45; p = 0.02) and showed a trend to longer disease-free survival when receiving adjuvant therapy (p = 0.09). CONCLUSION Our data support age as an independent negative prognostic factor in melanoma. Our data suggest that age does not affect primary surgical treatment but may affect decisions of whether or not patients receive postoperative treatment(s). Further work is needed to better understand the biological variables affecting treatment decisions and efficacy in older patients.
Collapse
Affiliation(s)
- Nathaniel H Fleming
- Interdisciplinary Melanoma Cooperative Group, New York University School of Medicine, New York, N.Y., USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fehres CM, Garcia-Vallejo JJ, Unger WWJ, van Kooyk Y. Skin-resident antigen-presenting cells: instruction manual for vaccine development. Front Immunol 2013; 4:157. [PMID: 23801994 PMCID: PMC3687254 DOI: 10.3389/fimmu.2013.00157] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/07/2013] [Indexed: 12/12/2022] Open
Abstract
The induction of antigen-specific effector T cells is driven by proper antigen presentation and co-stimulation by dendritic cells (DCs). For this reason strategies have been developed to instruct DCs for the induction of CD4+ and CD8+ T cell responses. Since DCs are localized, amongst other locations, in peripheral tissues such as the skin, new vaccines are aiming at targeting antigens to DCs in situ. Optimal skin-DC targeting in combination with adequate adjuvant delivery facilitates DC maturation and migration to draining lymph nodes and enhances antigen cross-presentation and T cell priming. In this review we describe what DC subsets populate the human skin, as well as current vaccination strategies based on targeting strategies and alternative administration for the induction of robust long-lived anti-cancer effector T cells.
Collapse
Affiliation(s)
- Cynthia M Fehres
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | | | | | | |
Collapse
|