1
|
Alla SSM, Tekuru Y, Lokesh MS, Alla D, Tvisha P, Tirupati S, Singh A, Tejaswini Y, Mahmood M, Siingh NP, Vineetha B. Talimogene laherparepvec (T-VEC) as a treatment for melanoma: A systematic review. J Oncol Pharm Pract 2025; 31:481-487. [PMID: 39819188 DOI: 10.1177/10781552241312920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Background and aimsMelanoma now presents an average risk of 1 in 50 in the Western world. Talimogene laherparepvec (T-VEC), an FDAapproved oncolytic virus derived from Herpes Simplex Virus type 1 (HSV-1), has proven effective in reducing morbidity and mortality from melanoma but causes adverse effects like chills, fever, exhaustion, and injection site discomfort. Research focuses on combining T-VEC with immune checkpoint inhibitors, such as pembrolizumab, to enhance its efficacy and broaden its application.MethodsA systematic search was conducted using PubMed, Scopus, Web of Science, Google Scholar, and ProMED, adhering to PRISMA guidelines. Results were tabulated and analyzed.ResultsThis review included 15 studies comprising nine cohorts, four case reports, a case series, and a randomized control trial, involving 779 melanoma patients in stages IIIB to IV, 58% of whom were male with a mean age of 65 years. Treatment duration with T-VEC averaged 35.07 weeks, with dosages ranging from 10^6 to 10^8 PFU/ml. The intervention yielded a mean DRR of 41.87% and an ORR of 62.2%. The most common side effect was chills, affecting 21.69% of participants. Pyrexia was reported by 20.41% of participants, followed by influenzalike illness (14.89%).ConclusionT-VEC effectively improves ORR and DRR in melanoma patients. However, further research is needed on combination therapy prospects and its adverse effects.
Collapse
Affiliation(s)
| | - Yogesh Tekuru
- R.V.M. Institute of Medical Sciences and Research Center, Telangana, India
| | - Moraboina Sai Lokesh
- Apollo Institute of Medical Sciences and Research Chittoor, Andhra Pradesh, India
| | - Deekshitha Alla
- Andhra Medical College, Visakhapatnam, Andhra Pradesh, India
| | - Patel Tvisha
- Government Medical College Surat, Gujarat, India
| | | | - Aradhya Singh
- Windsor University School of Medicine, Saint Kitts & Nevis
| | | | | | | | - Bodipudi Vineetha
- Apollo Institute of Medical Sciences and Research Chittoor, Andhra Pradesh, India
| |
Collapse
|
2
|
Sarangi R, Mishra S, Mahapatra S. Cancer Vaccines: A Novel Revolutionized Approach to Cancer Therapy. Indian J Clin Biochem 2025; 40:191-200. [PMID: 40123637 PMCID: PMC11928706 DOI: 10.1007/s12291-024-01201-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/19/2024] [Indexed: 03/25/2025]
Abstract
Over the past few decades, there has been significant advancement in the field of tumor immunotherapy. For many years vaccination against infectious diseases have been available. On the other hand very few cancer vaccines have been approved for human use. Ideal Cancer vaccines are biological response modifier work by stimulating both humoral and cellular immunity while overcoming the immunological suppression found in tumor. Two types of cancer vaccine: Prophylactic and therapeutic cancer vaccines are recommended for clinical use of individuals. HPV and HBV vaccines are the two widely used preventive vaccine used for treatment of cervical and hepatocellular carcinoma respectively and are approved by Food and Drug Administration (FDA). In therapeutic vaccine only three are approved: Sipuleucel T-cell vaccine for treatment refractory prostatic cancer, BCG vaccine for early bladder cancer and T-VEC for inoperable melanoma. Active ingredient in all cancer vaccines is an antigen. Antigens used for formulating cancer vaccines along with adjuvants optimizes immunogenicity in it. Heterogeneity within and between cancer types, screening and identifying suitable antigen specific to tumors and selection of vaccine delivery platforms are challenges in the development of vaccines. Adoptive cell therapy, Chimeric antigen receptor T cell therapy are recent breakthrough for cancer treatment.
Collapse
Affiliation(s)
- RajLaxmi Sarangi
- Departments of Biochemistry, Kalinga Institute of Medical Sciences (KIMS), Bhubaneswar, Odisha 751024 India
| | - Sanjukta Mishra
- Departments of Biochemistry, Kalinga Institute of Medical Sciences (KIMS), Bhubaneswar, Odisha 751024 India
| | - Srikrushna Mahapatra
- Departments of Biochemistry, Kalinga Institute of Medical Sciences (KIMS), Bhubaneswar, Odisha 751024 India
| |
Collapse
|
3
|
Timis T, Buruiana S, Dima D, Nistor M, Muresan XM, Cenariu D, Tigu AB, Tomuleasa C. Advances in Cell and Immune Therapies for Melanoma. Biomedicines 2025; 13:98. [PMID: 39857682 PMCID: PMC11761552 DOI: 10.3390/biomedicines13010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
The incidence rate of cutaneous melanoma is on the rise worldwide, due to increased exposure to UV radiation, aging populations, and exposure to teratogen agents. However, diagnosis is more precise, and the increased number of new cases is related to the improved diagnosis tools. Despite better early diagnosis and better therapies, melanoma has remained a significant public health challenge because of its aggressive behavior and high potential for metastasis. In 2020, cutaneous melanoma constituted approximately 1.3% of all cancer deaths that occurred within the European Union, thereby highlighting the necessity for effective prevention, timely diagnosis, and sustainable treatment measures, especially as a growing number of cases occur among younger patients. Melanoma is regarded as one of the most inflamed cancers due to its high immune cell presence and strong response to immunotherapy, fueling the need for development of immune-driven innovative treatments. Approved therapies, including immune checkpoint inhibitors (e.g., anti-PD-1 and anti-CTLA-4), have notably improved survival rates in melanoma. However, the limitations of the PD-1/PD-L1 and CTLA-4 axes inhibitors, such as low response rates, treatment resistance, and toxicity, have driven the need for continued research and advancements in treatment strategies. Current clinical trials are exploring various combinations of immune checkpoint inhibitors with costimulatory receptor agonists, chemotherapy, targeted therapies, and other immunotherapies, with the goal of improving outcomes and reducing side effects for melanoma patients. Emerging approaches, including adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) and oncolytic virotherapy, are showing promise. While CAR-T cell therapy has been less successful in melanoma compared to blood cancers, ongoing research is addressing challenges like the tumor microenvironment and antigen specificity. This review provides an overview of the requirement for advances in these medications, to mark a significant step forward in melanoma management, set to bring a fresh breath of hope for patients.
Collapse
Affiliation(s)
- Tanase Timis
- Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Department of Oncology, Bistrița Emergency Hospital, 420094 Bistrița, Romania
| | - Sanda Buruiana
- Department of Hematology, Nicolae Testemițanu University of Medicine and Pharmacy, MD-2004 Chisinau, Moldova
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Madalina Nistor
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ximena Maria Muresan
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Diana Cenariu
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Adrian-Bogdan Tigu
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
4
|
Gazal S, Olagnier D. Response to the letter to editor "Clarification on Rigvir's mechanism: Interpretation of CD55 inhibition in oncolytic therapy". Virology 2025; 601:110274. [PMID: 39532568 DOI: 10.1016/j.virol.2024.110274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Sabahat Gazal
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu, Jammu & Kashmir, India; Department of Biomedicine, Aarhus University, Denmark
| | | |
Collapse
|
5
|
Piskorz WM, Krętowski R, Cechowska-Pasko M. Marizomib Promotes Senescence or Long-Term Apoptosis in Melanoma Cancer Cells. Molecules 2024; 29:5652. [PMID: 39683813 DOI: 10.3390/molecules29235652] [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: 11/05/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Cellular senescence is cell cycle arrest and the inhibition of cell proliferation. New anticancer approaches include the elimination of cancer cells through the induction of senescence followed by senolysis. New prosenescence compounds are still being searched for. Little is known about the ability of proteasome inhibitors to induce senescence in tumor cells, especially in malignant melanoma. The aim of our study was to verify the activity of a natural proteasome inhibitor-marizomib (MZB)-directly after incubation and after its removal to assess its potential to induce senescence or long-term apoptosis in malignant melanoma cell lines (A375 and G361). After 48 h of incubation with MZB, we observed an increased number of SA-β-galactosidase-positive cells, upregulated expression of P21 and P-P53 proteins and an increased number of cells at the subG1 phase (line G361) or at both the subG1 and G2/M phases (line A375). After 96 h from inhibitor removal, the G361 line presented signs of senescence (increased level of SA-β-galactosidase, IL-8, P-P53, G2/M and S phases of cell cycle, decreased lamin B1 and cleaved lamin B1), while the A375 line demonstrated more signs of apoptosis (increased subG1 phase, P-P53, cleaved lamin B1). The gathered findings suggest that MZB resulted in the induction of cellular senescence (line G361) or enhanced apoptosis (line A375) in the melanoma cell lines tested here and could be a promising therapeutic factor in malignant melanoma treatment.
Collapse
Affiliation(s)
- Wiktoria Monika Piskorz
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Białystok, Poland
| | - Rafał Krętowski
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Białystok, Poland
| | - Marzanna Cechowska-Pasko
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Białystok, Poland
| |
Collapse
|
6
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
7
|
Jimenez S, Powell RP, Rincon L, Clement C. A 64-Year-Old Woman With Molluscum Contagiosum Arising in a Melanocytic Nevus: A Case Report of an Unusual Coexistence. Cureus 2024; 16:e71646. [PMID: 39553005 PMCID: PMC11567655 DOI: 10.7759/cureus.71646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Molluscum contagiosum (MC) virus is a poxvirus that infects epidermal keratinocytes producing cutaneous nodules with characteristic intracytoplasmic inclusions. Intradermal nevus is a benign lesion that typically presents as clusters of melanocytes within the dermal layer of skin. Although both MC and melanocytic nevus are common lesions, MC arising within a melanocytic nevus is a rare event with only a few cases published in the literature. Herein, we report the case of a 64-year-old immunocompetent woman with an MC emerging within an intradermal nevus of the left nasolabial fold. In addition, the disappearance of most dermal melanocytes adjacent to the site of the MC infection was noted. This is a rarely reported event, and it has been suggested that it involves a mechanism of viral oncolysis among others, with potential implications for treatment and a need for further investigation.
Collapse
Affiliation(s)
- Sylvia Jimenez
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, USA
| | - Reuben P Powell
- Pathology, University of Texas Medical Branch, Galveston, USA
| | - Liliana Rincon
- Pathology, University of Texas Medical Branch, Galveston, USA
| | - Cecilia Clement
- Pathology, University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
8
|
Antonova L, Paramanthan P, Falls T, Wedge ME, Mayer J, Sekhon HS, McPherson J, Denroche RE, Gallinger S, Bell JC, Ilkow CS, Chatterjee A. Molecular Characterization and Xenotransplantation of Pancreatic Cancer Using Endoscopic Ultrasound-Guided Fine Needle Aspiration (EUS-FNA). Cancers (Basel) 2024; 16:2721. [PMID: 39123450 PMCID: PMC11311391 DOI: 10.3390/cancers16152721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Pancreatic cancer has one of the worst prognoses among all malignancies and few available treatment options. Patient-derived xenografts can be used to develop personalized therapy for pancreatic cancer. Endoscopic ultrasound fine-needle aspiration (EUS-FNA) may provide a powerful alternative to surgery for obtaining sufficient tissue for the establishment of patient-derived xenografts. In this study, EUS-FNA samples were obtained for 30 patients referred to the Ottawa Hospital, Ottawa, Ontario, Canada. These samples were used for xenotransplantation in NOD-SCID mice and for genetic analyses. The gene expression of pancreatic-cancer-relevant genes in xenograft tumors was examined by immunohistochemistry. Targeted sequencing of both the patient-derived tumors and xenograft tumors was performed. The xenografts' susceptibility to oncolytic virus infection was studied by infecting xenograft-derived cells with VSV∆51-GFP. The xenograft take rate was found to be 75.9% for passage 1 and 100% for passage 2. Eighty percent of patient tumor samples were successfully sequenced to a high depth for 42 cancer genes. Xenograft histological characteristics and marker expression were maintained between passages. All tested xenograft samples were susceptible to oncoviral infection. We found that EUS-FNA is an accessible, minimally invasive technique that can be used to acquire adequate pancreatic cancer tissue for the generation of patient-derived xenografts and for genetic sequencing.
Collapse
Affiliation(s)
- Lilia Antonova
- Department of Otolaryngology-Head and Neck Surgery, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Piriya Paramanthan
- Division of Gastroenterology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Theresa Falls
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Marie-Eve Wedge
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Justin Mayer
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Harman S. Sekhon
- Division of Anatomic Pathology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada;
| | - John McPherson
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | | | - Steven Gallinger
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - John Cameron Bell
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Carolina S. Ilkow
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Avijit Chatterjee
- Division of Gastroenterology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
9
|
Slominski RM, Kim TK, Janjetovic Z, Brożyna AA, Podgorska E, Dixon KM, Mason RS, Tuckey RC, Sharma R, Crossman DK, Elmets C, Raman C, Jetten AM, Indra AK, Slominski AT. Malignant Melanoma: An Overview, New Perspectives, and Vitamin D Signaling. Cancers (Basel) 2024; 16:2262. [PMID: 38927967 PMCID: PMC11201527 DOI: 10.3390/cancers16122262] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Melanoma, originating through malignant transformation of melanin-producing melanocytes, is a formidable malignancy, characterized by local invasiveness, recurrence, early metastasis, resistance to therapy, and a high mortality rate. This review discusses etiologic and risk factors for melanoma, diagnostic and prognostic tools, including recent advances in molecular biology, omics, and bioinformatics, and provides an overview of its therapy. Since the incidence of melanoma is rising and mortality remains unacceptably high, we discuss its inherent properties, including melanogenesis, that make this disease resilient to treatment and propose to use AI to solve the above complex and multidimensional problems. We provide an overview on vitamin D and its anticancerogenic properties, and report recent advances in this field that can provide solutions for the prevention and/or therapy of melanoma. Experimental papers and clinicopathological studies on the role of vitamin D status and signaling pathways initiated by its active metabolites in melanoma prognosis and therapy are reviewed. We conclude that vitamin D signaling, defined by specific nuclear receptors and selective activation by specific vitamin D hydroxyderivatives, can provide a benefit for new or existing therapeutic approaches. We propose to target vitamin D signaling with the use of computational biology and AI tools to provide a solution to the melanoma problem.
Collapse
Affiliation(s)
- Radomir M. Slominski
- Department of Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Tae-Kang Kim
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Zorica Janjetovic
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Ewa Podgorska
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Katie M. Dixon
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Rebecca S. Mason
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Robert C. Tuckey
- School of Molecular Sciences, University of Western Australia, Perth, WA 6009, Australia;
| | - Rahul Sharma
- Department of Biomedical Informatics and Data Science, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - David K. Crossman
- Department of Genetics and Bioinformatics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Craig Elmets
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Chander Raman
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anton M. Jetten
- Cell Biology Section, NIEHS—National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrzej T. Slominski
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, Veteran Administration Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
10
|
Wang C, Lu N, Yan L, Li Y. The efficacy and safety assessment of oncolytic virotherapies in the treatment of advanced melanoma: a systematic review and meta-analysis. Virol J 2023; 20:252. [PMID: 37919738 PMCID: PMC10623758 DOI: 10.1186/s12985-023-02220-x] [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: 01/15/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND The efficacy and safety of oncolytic virotherapies in the treatment of advanced melanoma still remains controversal. It is necessary to conduct quantitative evaluation on the basis of preclinical trial reports. METHODS Publicly available databases (PubMed, Embase, Medline, Web of Science and Cochrane Library.) and register (Clinicaltrials.gov) were searched to collect treatment outcomes of oncolytic virotherapies (including herpes simplex virus type 1 (HSV), coxsackievirus A21 (CVA21), adenovirus, poxvirus and reovirus) for advanced/unresectable melanoma. Comparisons of treatment response, adverse events (AEs) and survival analyses for different virotherapies were performed by R software based on the extracted data from eligible studies. RESULTS Finally, thirty-four eligible studies were analysed and HSV virotherapy had the highest average complete response (CR, 24.8%) and HSV had a slightly higher average overall response rate (ORR) than CVA21 (43.8% vs 42.6%). In the pooled results of comparing talimogene laherparepve (T-VEC) with or without GM-CSF/ICIs (immune checkpoint inhibitors) to GM-CSF/ICIs monotherapy suggested virotherapy was more efficient in subgroups CR (RR = 1.80, 95% CI [1.30; 2.51], P < 0.01), ORR (RR = 1.17, 95% CI [1.02; 1.34], P < 0.05), and DCR (RR = 1.27, 95% CI [1.15; 1.40], P < 0.01). In patients treated with T-VEC+ICIs, 2-year overall survival (12.1 ± 6.9 months) and progression-free survival (9.9 ± 6.9) were significantly longer than those treated with T-VEC alone. Furthermore, we found that AEs occurred frequently in virotherapy but decreased in a large cohort of enrolled patients, some of which, such as abdominal distension/pain, injection site pain and pruritus, were found to be positively associated with disease progression in patients treated with T-VEC monotherapy. CONCLUSION Given the relative safety and tolerability of oncolytic viruses, and the lack of reports of dose-limiting-dependent toxicities, more patients treated with T-VEC with or without ICIs should be added to future assessment analyses. There is still a long way to go before it can be used as a first-line therapy for patients with advanced or unresectable melanoma.
Collapse
Affiliation(s)
- Changyuan Wang
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China
| | - Nanxiao Lu
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China
| | - Lin Yan
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China
| | - Yang Li
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China.
| |
Collapse
|
11
|
Huang Q, Pan X, Zhu W, Zhao W, Xu H, Hu K. Natural Products for the Immunotherapy of Glioma. Nutrients 2023; 15:2795. [PMID: 37375698 DOI: 10.3390/nu15122795] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Glioma immunotherapy has attracted increasing attention since the immune system plays a vital role in suppressing tumor growth. Immunotherapy strategies are already being tested in clinical trials, such as immune checkpoint inhibitors (ICIs), vaccines, chimeric antigen receptor T-cell (CAR-T cell) therapy, and virus therapy. However, the clinical application of these immunotherapies is limited due to their tremendous side effects and slight efficacy caused by glioma heterogeneity, antigen escape, and the presence of glioma immunosuppressive microenvironment (GIME). Natural products have emerged as a promising and safe strategy for glioma therapy since most of them possess excellent antitumor effects and immunoregulatory properties by reversing GIME. This review summarizes the status of current immunotherapy strategies for glioma, including their obstacles. Then we discuss the recent advancement of natural products for glioma immunotherapy. Additionally, perspectives on the challenges and opportunities of natural compounds for modulating the glioma microenvironment are also illustrated.
Collapse
Affiliation(s)
- Qi Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xier Pan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhao Zhu
- Department of Anaesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wen Zhao
- Department of Anaesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hongzhi Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
- Neurosurgical Institute, Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
12
|
Ruiz EM, Alhassan SA, Errami Y, Abd Elmageed ZY, Fang JS, Wang G, Brooks MA, Abi-Rached JA, Kandil E, Zerfaoui M. A Predictive Model of Adaptive Resistance to BRAF/MEK Inhibitors in Melanoma. Int J Mol Sci 2023; 24:8407. [PMID: 37176114 PMCID: PMC10178962 DOI: 10.3390/ijms24098407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The adaptive acquisition of resistance to BRAF and MEK inhibitor-based therapy is a common feature of melanoma cells and contributes to poor patient treatment outcomes. Leveraging insights from a proteomic study and publicly available transcriptomic data, we evaluated the predictive capacity of a gene panel corresponding to proteins differentially abundant between treatment-sensitive and treatment-resistant cell lines, deciphering predictors of treatment resistance and potential resistance mechanisms to BRAF/MEK inhibitor therapy in patient biopsy samples. From our analysis, a 13-gene signature panel, in both test and validation datasets, could identify treatment-resistant or progressed melanoma cases with an accuracy and sensitivity of over 70%. The dysregulation of HMOX1, ICAM, MMP2, and SPARC defined a BRAF/MEK treatment-resistant landscape, with resistant cases showing a >2-fold risk of expression of these genes. Furthermore, we utilized a combination of functional enrichment- and gene expression-derived scores to model and identify pathways, such as HMOX1-mediated mitochondrial stress response, as potential key drivers of the emergence of a BRAF/MEK inhibitor-resistant state in melanoma cells. Overall, our results highlight the utility of these genes in predicting treatment outcomes and the underlying mechanisms that can be targeted to reduce the development of resistance to BRAF/MEK targeted therapy.
Collapse
Affiliation(s)
- Emmanuelle M. Ruiz
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Solomon A. Alhassan
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Youssef Errami
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Zakaria Y. Abd Elmageed
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of Louisiana, Monroe, LA 71203, USA
| | - Jennifer S. Fang
- Department of Cell and Molecular Biology, Tulane University School of Science & Engineering, New Orleans, LA 70118, USA
| | - Guangdi Wang
- Department of Chemistry, RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Margaret A. Brooks
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Joe A. Abi-Rached
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Emad Kandil
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Mourad Zerfaoui
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
13
|
Anestopoulos I, Kyriakou S, Tragkola V, Paraskevaidis I, Tzika E, Mitsiogianni M, Deligiorgi MV, Petrakis G, Trafalis DT, Botaitis S, Giatromanolaki A, Koukourakis MI, Franco R, Pappa A, Panayiotidis MI. Targeting the epigenome in malignant melanoma: Facts, challenges and therapeutic promises. Pharmacol Ther 2022; 240:108301. [PMID: 36283453 DOI: 10.1016/j.pharmthera.2022.108301] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
Malignant melanoma is the most lethal type of skin cancer with high rates of mortality. Although current treatment options provide a short-clinical benefit, acquired-drug resistance highlights the low 5-year survival rate among patients with advanced stage of the disease. In parallel, the involvement of an aberrant epigenetic landscape, (e.g., alterations in DNA methylation patterns, histone modifications marks and expression of non-coding RNAs), in addition to the genetic background, has been also associated with the onset and progression of melanoma. In this review article, we report on current therapeutic options in melanoma treatment with a focus on distinct epigenetic alterations and how their reversal, by specific drug compounds, can restore a normal phenotype. In particular, we concentrate on how single and/or combinatorial therapeutic approaches have utilized epigenetic drug compounds in being effective against malignant melanoma. Finally, the role of deregulated epigenetic mechanisms in promoting drug resistance to targeted therapies and immune checkpoint inhibitors is presented leading to the development of newly synthesized and/or improved drug compounds capable of targeting the epigenome of malignant melanoma.
Collapse
Affiliation(s)
- I Anestopoulos
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - S Kyriakou
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - V Tragkola
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - I Paraskevaidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - E Tzika
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | | | - M V Deligiorgi
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - G Petrakis
- Saint George Hospital, Chania, Crete, Greece
| | - D T Trafalis
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - S Botaitis
- Department of Surgery, Alexandroupolis University Hospital, Democritus University of Thrace School of Medicine, Alexandroupolis, Greece
| | - A Giatromanolaki
- Department of Pathology, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - M I Koukourakis
- Radiotherapy / Oncology, Radiobiology & Radiopathology Unit, Department of Medicine, School of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - R Franco
- Redox Biology Centre, University of Nebraska-Lincoln, Lincoln, NE, USA; School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - A Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - M I Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.
| |
Collapse
|
14
|
Gartrell RD, Blake Z, Rizk EM, Perez-Lorenzo R, Weisberg SP, Simoes I, Esancy C, Fu Y, Davari DR, Barker L, Finkel G, Mondal M, Minns HE, Wang SW, Fullerton BT, Lozano F, Chiuzan C, Horst B, Saenger YM. Combination immunotherapy including OncoVEX mGMCSF creates a favorable tumor immune micro-environment in transgenic BRAF murine melanoma. Cancer Immunol Immunother 2022; 71:1837-1849. [PMID: 34999916 PMCID: PMC10991384 DOI: 10.1007/s00262-021-03088-y] [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: 06/08/2021] [Accepted: 10/05/2021] [Indexed: 12/01/2022]
Abstract
Talimogene Laherparepvec (OncoVEXmGMCSF), an oncolytic virus, immune checkpoint inhibitor anti-programmed cell death protein 1 (anti-PD1), and BRAF inhibition (BRAFi), are all clinically approved for treatment of melanoma patients and are effective through diverse mechanisms of action. Individually, these therapies also have an effect on the tumor immune microenvironment (TIME). Evaluating the combination effect of these three therapies on the TIME can help determine when combination therapy is most appropriate for further study. In this study, we use a transgenic murine melanoma model (Tyr::CreER; BRAFCA/+; PTENflox/flox), to evaluate the TIME in response to combinations of BRAFi, anti-PD1, and OncoVEXmGMCSF. We find that mice treated with the triple combination BRAFi + anti-PD1 + OncoVEXmGMCSF have decreased tumor growth compared to BRAFi alone and prolonged survival compared to control. Flow cytometry shows an increase in percent CD8 + /CD3 + cytotoxic T Lymphocytes (CTLs) and a decrease in percent FOXP3 + /CD4 + T regulatory cells (Tregs) in tumors treated with OncoVEXmGMCSF compared to mice not treated with OncoVEXmGMCSF. Immunogenomic analysis at 30d post-treatment shows an increase in Th1 and interferon-related genes in mice receiving OncoVEXmGMCSF + BRAFi. In summary, treatment with combination BRAFi + anti-PD1 + OncoVEXmGMCSF is more effective than any single treatment in controlling tumor growth, and groups receiving OncoVEXmGMCSF had more tumoral infiltration of CTLs and less intratumoral Tregs in the TIME. This study provides rational basis to combine targeted agents, oncolytic viral therapy, and checkpoint inhibitors in the treatment of melanoma.
Collapse
Affiliation(s)
- Robyn D Gartrell
- Department of Pediatrics, Columbia University Irving Medical Center, 1130 St. Nicholas Avenue, ICRC 916A, New York, NY, 10032, USA
| | - Zoë Blake
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, PS 9-428, New York, NY, 10032, USA
| | - Emanuelle M Rizk
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, PS 9-428, New York, NY, 10032, USA
| | - Rolando Perez-Lorenzo
- Department of Dermatology, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Russ Berrie Medical Science Pavillion Room 307, New York, NY, 10032, USA
| | - Stuart P Weisberg
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Ines Simoes
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut d'Investigacions Biomediques August Pi I Sunyer, Barcelona, Catalunya, Spain
| | - Camden Esancy
- Herbert Irving Comprehensicve Cancer Center, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Yichun Fu
- Department of Medicine, Mount Sinai Hospital, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Danielle R Davari
- University of North Carolina School of Medicine, 140 W Franklin Street, Unit 506, Chapel Hill, NC, 27516, USA
| | - Luke Barker
- Valegos College of Physicians and Surgeons, Columbia University, 630 W 168th Street, New York, NY, 10032, USA
| | - Grace Finkel
- Valegos College of Physicians and Surgeons, Columbia University, 630 W 168th Street, New York, NY, 10032, USA
| | - Manas Mondal
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, PS 9-428, New York, NY, 10032, USA
| | - Hanna E Minns
- Department of Pediatrics, Columbia University Irving Medical Center, 1130 St. Nicholas Avenue, ICRC 916A, New York, NY, 10032, USA
| | - Samuel W Wang
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, PS 9-428, New York, NY, 10032, USA
| | - Benjamin T Fullerton
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, PS 9-428, New York, NY, 10032, USA
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut d'Investigacions Biomediques August Pi I Sunyer, Barcelona, Catalunya, Spain
- Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Codruta Chiuzan
- Department of Biostatistics, Columbia University Irving Medical Center, 722 W 168th Street, Room 646, New York, NY, 10032, USA
| | - Basil Horst
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Yvonne M Saenger
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, PS 9-428, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Revisiting the melanomagenic pathways and current therapeutic approaches. Mol Biol Rep 2022; 49:9651-9671. [DOI: 10.1007/s11033-022-07412-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
|
16
|
Aboul-Fettouh N, Morse D, Patel J, Migden MR. Immunotherapy and Systemic Treatment of Cutaneous Squamous Cell Carcinoma. Dermatol Pract Concept 2021; 11:e2021169S. [PMID: 34877077 DOI: 10.5826/dpc.11s2a169s] [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] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Cutaneous squamous cell carcinomas (cSCC) represent one of the most diagnosed non-melanoma skin cancers and its incidence is increasing globally. Whereas early stage and low risk cSCC is typically treated with surgery, and in some cases other localized therapeutic modalities, locally advanced or metastatic cSCC is a cause of significant morbidity and mortality that requires a different approach to therapy. Therapeutic attempts at treating advanced cSCC include a multi-disciplinary approach with considerations for surgery, radiation, and systemic therapies. In this review, we will discuss the various systemic therapies that have been trialed for advanced cSCC, beginning with the early cytotoxic and platinum-based agents as well as their corresponding limitations. We will then review the targeted approaches using EGFR inhibitors prior to discussing the more recent immunotherapeutics that have shown good tumor responses in this often-lethal disease.
Collapse
Affiliation(s)
- Nader Aboul-Fettouh
- Department of Dermatology, The University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Daniel Morse
- Department of Dermatology, The University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Jigar Patel
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael R Migden
- Departments of Dermatology and Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
O'Connell F, O'Sullivan J. Help or hindrance: The obesity paradox in cancer treatment response. Cancer Lett 2021; 522:269-280. [PMID: 34534616 DOI: 10.1016/j.canlet.2021.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
Obesity is a rising epidemic, the influence of which on cancer development, progression as well as its impact on current standard of care cancer treatments is profound with many facets. Obesity is emerging as a modulating factor in many cancer therapies, such as chemotherapy, radiotherapy, immunotherapy and combination therapies. It has been reported to diminish the efficacy of some treatments but has also been alluded to being protective in terms of reduced treatment toxicities, thus the evolution of the obesity paradox. The obese tumour microenvironment influences treatment response through modulation of a series of aspects, including altered adipocyte secretome, angiogenesis, hypoxia, fibrosis, free fatty acid uptake as well as a modulated immune landscape. However, the influence of these underlying mechanisms on cancer treatment response and the biological action of adipose tissue is still largely unknown. Elucidation of these facets may lead to the enhanced efficacy of current treatment options or the identification of novel methods to combat cancer in the obese tumour microenvironment.
Collapse
Affiliation(s)
- Fiona O'Connell
- Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Jacintha O'Sullivan
- Trinity St. James's Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
18
|
Salloum A, Koblinski J, Bazzi N, Zeitouni NC. Talimogene Laherparepvec in Non-Melanoma Cancers. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2021; 14:18-25. [PMID: 34980955 PMCID: PMC8675341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND Talimogene laherparepvec (T-VEC) is the first oncolytic virus therapy approved by the United States Food and Drug Administration (in 2015) for the treatment of advanced-stage melanoma. Despite a paucity of Phase III trials for T-VEC as a therapy for non-melanoma cancers, successful off-label use of T-VEC for this purpose has been reported in the literature. OBJECTIVE We sought to review the literature describing T-VEC as a treatment for non-melanoma cancer. METHODS Systematic searches of the PubMed literature database and ClinicalTrials.gov website were performed in July 2020, focusing on T-VEC in combination with non-melanoma cancer, including squamous cell carcinoma, Merkel cell carcinoma, sarcoma, cutaneous B-cell lymphoma, and cutaneous T-cell lymphoma. Articles were screened based on their title and abstract. RESULTS Nine articles with 87 patients were included. Relevant articles included case reports, case series, and Phase I and Phase II trials. The majority of patients in the studies had refractory cancers or had been heavily pretreated. Overall, T-VEC demonstrated efficacy for non-melanoma cancer, both independently and in combination with biologics. CONCLUSION T-VEC has demonstrated efficacy for non-melanoma cancers. Phase III trials of T-VEC for this indication are warranted to expand its clinical utility.
Collapse
Affiliation(s)
- Antoine Salloum
- Dr. Salloum is with the Department of Dermatology at Saint George Hospital University Medical Center in Beirut, Lebanon, and Dermatologic Surgicenter in Philadelphia, Pennsylvania
- Drs. Koblinski and Zeitouni are with the University of Arizona College of Medicine in Phoenix, Arizona
- Dr. Bazzi is with the Faculty of Medicine at Lebanese University in Beirut, Lebanon
| | - Jenna Koblinski
- Dr. Salloum is with the Department of Dermatology at Saint George Hospital University Medical Center in Beirut, Lebanon, and Dermatologic Surgicenter in Philadelphia, Pennsylvania
- Drs. Koblinski and Zeitouni are with the University of Arizona College of Medicine in Phoenix, Arizona
- Dr. Bazzi is with the Faculty of Medicine at Lebanese University in Beirut, Lebanon
| | - Nagham Bazzi
- Dr. Salloum is with the Department of Dermatology at Saint George Hospital University Medical Center in Beirut, Lebanon, and Dermatologic Surgicenter in Philadelphia, Pennsylvania
- Drs. Koblinski and Zeitouni are with the University of Arizona College of Medicine in Phoenix, Arizona
- Dr. Bazzi is with the Faculty of Medicine at Lebanese University in Beirut, Lebanon
| | - Nathalie C Zeitouni
- Dr. Salloum is with the Department of Dermatology at Saint George Hospital University Medical Center in Beirut, Lebanon, and Dermatologic Surgicenter in Philadelphia, Pennsylvania
- Drs. Koblinski and Zeitouni are with the University of Arizona College of Medicine in Phoenix, Arizona
- Dr. Bazzi is with the Faculty of Medicine at Lebanese University in Beirut, Lebanon
| |
Collapse
|
19
|
Oncolytic Virotherapy for Melanoma Brain Metastases, a Potential New Treatment Paradigm? Brain Sci 2021; 11:brainsci11101260. [PMID: 34679325 PMCID: PMC8534242 DOI: 10.3390/brainsci11101260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Melanoma brain metastases remain a devastating disease process with poor prognosis. Recently, there has been a surge in studies demonstrating the efficacy of oncolytic virotherapy for brain tumor treatment. Given their specificity and amenability to genetic modification, the authors explore the possible role of oncolytic virotherapy as a potential treatment option for patients with melanoma brain metastases. METHODS A comprehensive literature review including both preclinical and clinical evidence of oncolytic virotherapy for the treatment of melanoma brain metastasis was performed. RESULTS Oncolytic virotherapy, specifically T-VEC (Imlygic™), was approved for the treatment of melanoma in 2015. Recent clinical trials demonstrate promising anti-tumor changes in patients who have received T-VEC; however, there is little evidence for its use in metastatic brain disease based on the existing literature. To date, only two single cases utilizing virotherapy in patients with metastatic brain melanoma have been reported, specifically in patients with treatment refractory disease. Currently, there is not sufficient data to support the use of T-VEC or other viruses for intracranial metastatic melanoma. In developing a virotherapy treatment paradigm for melanoma brain metastases, several factors must be considered, including route of administration, need to bypass the blood-brain barrier, viral tumor infectivity, and risk of adverse events. CONCLUSIONS Evidence for oncolytic virotherapy treatment of melanoma is limited primarily to T-VEC, with a noticeable paucity of data in the literature with respect to brain tumor metastasis. Given the promising findings of virotherapy for other brain tumor types, oncolytic virotherapy has great potential to offer benefits to patients afflicted with melanoma brain metastases and warrants further investigation.
Collapse
|
20
|
Current understanding of nonsurgical interventions for refractory differentiated thyroid cancer: a systematic review. Future Sci OA 2021; 7:FSO738. [PMID: 34258030 PMCID: PMC8256328 DOI: 10.2144/fsoa-2021-0041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 12/28/2022] Open
Abstract
Thyroid cancer incidence and related mortality is increasing year-on-year, and although treatment for early disease with surgery and radioiodine results in a 98% 5-year survival rate, recurrence and treatment refractory disease is evident in an unacceptable number of patients. Alternative treatment regimens have therefore been sought in the form of tyrosine kinase inhibitors, immunotherapy, vaccines, chimeric antigen receptor T-cell therapy and oncolytic viruses. The current review aims to consolidate knowledge and highlight the latest clinical trials using secondary therapies in thyroid cancer treatment, focusing on both in vitro and in vivo studies, which have investigated therapies other than radioiodine. The rates of thyroid cancer and related deaths are increasing. Differentiated thyroid cancer is the most common type of thyroid cancer. Early disease can be treated with surgery and radioactive iodine with very good outcomes. However, this therapy does not work for a small number of patients making it important to find different (secondary) treatment options. This review summarizes the results of published research about secondary treatment options for differentiated thyroid cancer. Ongoing research including laboratory-based and clinical trials are also highlighted.
Collapse
|
21
|
MacArthur TA, Fahy AS, Jakub JW. Surgical Resection After Talimogene Laherparepvec for Melanoma: Persistent Fuorodeoxyglucose Avidity on Positron Emission Tomography Despite No Viable Disease. Am Surg 2021; 87:849-854. [PMID: 34060947 DOI: 10.1177/00031348211023434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Talimogene laherparepvec (TVEC) is an injectable attenuated oncolytic herpes simplex virus (HSV-1) used in the treatment of loco regionally metastatic melanoma. Lesions managed by TVEC are generally considered unresectable at time of initiation of intralesional therapy; however, a subset of patients go on to have surgical resection of loco regionally controlled disease. We sought to review our experience with surgical excision of treated lesions to offer an insight into the radiologic correlate, treatment effect, and pathological findings of intralesional TVEC therapy. METHODS This is a retrospective descriptive case series of patients who underwent TVEC injection at Mayo Clinic, Rochester, MN, between October 2016 and July 2020. Institutional Institutional Review Board approval was obtained. RESULTS Twenty-one patients underwent intralesional TVEC, met inclusion criteria, and were included in this series. Seven went on to surgical excision of the injected lesions after an initial course of TVEC. Of those 7 patients, 4 had residual melanoma in the specimen on final pathology, while 3 had a complete pathologic response. All 3 patients who had no residual disease on pathology continued to have fluorodeoxyglucose (FDG) avidity on preoperative positron emission tomography scan of the excised lesions. DISCUSSION Despite ongoing FDG avidity on PET scan, patients who have well-controlled disease and stability over time of the injected lesions may benefit from surgical excision following a course of TVEC. This may render the patient clinically disease free and/or allow them a reprieve from TVEC treatment.
Collapse
Affiliation(s)
| | | | - James W Jakub
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Han D, van Akkooi ACJ, Straker RJ, Shannon AB, Karakousis GC, Wang L, Kim KB, Reintgen D. Current management of melanoma patients with nodal metastases. Clin Exp Metastasis 2021; 39:181-199. [PMID: 33961168 PMCID: PMC8102663 DOI: 10.1007/s10585-021-10099-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/22/2021] [Indexed: 12/26/2022]
Abstract
The management of melanoma patients with nodal metastases has undergone dramatic changes over the last decade. In the past, the standard of care for patients with a positive sentinel lymph node biopsy (SLNB) was a completion lymph node dissection (CLND), while patients with palpable macroscopic nodal disease underwent a therapeutic lymphadenectomy in cases with no evidence of systemic spread. However, studies have shown that SLN metastases present as a spectrum of disease, with certain SLN-based factors being prognostic of and correlated with outcomes. Furthermore, the results of key clinical trials demonstrate that CLND provides no survival benefit over nodal observation in positive SLN patients, while other clinical trials have shown that adjuvant immune checkpoint inhibitor therapy or targeted therapy after CLND is associated with a recurrence-free survival benefit. Given the efficacy of these systemic therapies in the adjuvant setting, these agents are now being evaluated and utilized as neoadjuvant treatments in patients with regionally-localized or resectable metastatic melanoma. Multiple options now exist to treat melanoma patients with nodal disease, and determining the best treatment course for a particular case requires an in-depth knowledge of current data and an informed discussion with the patient. This review will provide an overview of the various options for treating melanoma patients with nodal metastases and will discuss the data that supported the development of these treatment options.
Collapse
Affiliation(s)
- Dale Han
- Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code: L619, Portland, OR, 97239, USA.
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Richard J Straker
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Adrienne B Shannon
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Giorgos C Karakousis
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Lin Wang
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | - Kevin B Kim
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | - Douglas Reintgen
- Department of Surgery, Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
23
|
Ballotti R, Cheli Y, Bertolotto C. The complex relationship between MITF and the immune system: a Melanoma ImmunoTherapy (response) Factor? Mol Cancer 2020; 19:170. [PMID: 33276788 PMCID: PMC7718690 DOI: 10.1186/s12943-020-01290-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
The clinical benefit of immune checkpoint inhibitory therapy (ICT) in advanced melanomas is limited by primary and acquired resistance. The molecular determinants of the resistance have been extensively studied, but these discoveries have not yet been translated into therapeutic benefits. As such, a paradigm shift in melanoma treatment, to surmount the therapeutic impasses linked to the resistance, is an important ongoing challenge.This review outlines the multifaceted interplay between microphthalmia-associated transcription factor (MITF), a major determinant of the biology of melanoma cells, and the immune system. In melanomas, MITF functions downstream oncogenic pathways and microenvironment stimuli that restrain the immune responses. We highlight how MITF, by controlling differentiation and genome integrity, may regulate melanoma-specific antigen expression by interfering with the endolysosomal pathway, KARS1, and antigen processing and presentation. MITF also modulates the expression of coinhibitory receptors, i.e., PD-L1 and HVEM, and the production of an inflammatory secretome, which directly affects the infiltration and/or activation of the immune cells.Furthermore, MITF is also a key determinant of melanoma cell plasticity and tumor heterogeneity, which are undoubtedly one of the major hurdles for an effective immunotherapy. Finally, we briefly discuss the role of MITF in kidney cancer, where it also plays a key role, and in immune cells, establishing MITF as a central mediator in the regulation of immune responses in melanoma and other cancers.We propose that a better understanding of MITF and immune system intersections could help in the tailoring of current ICT in melanomas and pave the way for clinical benefits and long-lasting responses.
Collapse
Affiliation(s)
- Robert Ballotti
- Université Côte d'Azur, Nice, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Yann Cheli
- Université Côte d'Azur, Nice, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Corine Bertolotto
- Université Côte d'Azur, Nice, France.
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France.
| |
Collapse
|
24
|
Audrey-Bayan C, Trager MH, Gartrell-Corrado RD, Rizk EM, Pradhan J, Silverman AM, Lopez A, Marks DK, Niedt G, Geskin LJ, Saenger YM. Distinguishing melanophages from tumor in melanoma patients treated with talimogene laherparepvec. Melanoma Res 2020; 30:410-415. [PMID: 32379409 PMCID: PMC10961165 DOI: 10.1097/cmr.0000000000000661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Response to talimogene laherparepvec (T-Vec) is difficult to assess as pigmented macrophages that have ingested melanoma cells ('melanophages') persist after injection, mimicking melanoma. We used quantitative immunofluorescence (qIF) to (1) distinguish melanophages from melanoma in biopsies from two patients treated with T-Vec and (2) evaluate the tumor microenvironment pretreatment and posttreatment. Tissues were stained with 4',6-diamidino-2-phenylindole, cluster of differentiation (CD) 3, CD8, CD68, human leukocyte antigen-DR isotype (HLA-DR), and SRY-Box Transcription Factor 10 (SOX10), and multispectral images were analyzed. Post-T-Vec samples showed melanophages with cytoplasmic costaining of CD68, SOX10, and HLA-DR, without nuclear SOX10 expression. qIF revealed a dense immune infiltrate of CD3, CD8, and CD68 cells in post-T-Vec samples. Melanophages from tumors post-T-Vec stain the nuclear melanoma marker SOX10 in their cytoplasms as compared to melanoma cells that stain nuclear SOX10. This novel finding highlights the phagocytosis of melanoma cell components by macrophages after treatment with T-Vec. qIF may assist pathologists in determining whether lesions treated with immunotherapy contain residual viable melanoma.
Collapse
Affiliation(s)
- Claire Audrey-Bayan
- Department of Medicine and Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Megan H. Trager
- Department of Dermatology, Division of Pediatric Hematology/Oncology
| | | | - Emanuelle M. Rizk
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center
| | - Jaya Pradhan
- Department of Pathology, Columbia University Irving Medical Center
| | | | - Adriana Lopez
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | - George Niedt
- Department of Dermatopathology, Columbia University Irving Medical Center, New York, New York, USA
| | - Larisa J. Geskin
- Department of Dermatology, Division of Pediatric Hematology/Oncology
| | - Yvonne M. Saenger
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center
| |
Collapse
|
25
|
Sorokins V, Tilgase A, Olmane E, Isajevs S, Zablocka T, Rasa A, Alberts P. A progressive stage IIIB melanoma treated with oncolytic ECHO-7 virus: A case report. SAGE Open Med Case Rep 2020; 8:2050313X20934978. [PMID: 32637110 PMCID: PMC7313323 DOI: 10.1177/2050313x20934978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 05/25/2020] [Indexed: 12/04/2022] Open
Abstract
Melanoma is an aggressive skin cancer form with a grave prognosis. Current results suggest that oncolytic virus treatment of melanoma has a high therapeutic potential. ECHO-7 (Rigvir) is the first oncolytic virus registered in Latvia. A female patient was diagnosed with stage IIIB melanoma in December 2012, over 9.4 years ago. After the first excision and re-excision, the patient had several recurrences and disease progressions. After the patient had received surgical treatment in 2014, ECHO-7 virus oncolytic virotherapy was started. Since then, the patient has experienced only one more disease progression episode in May 2015 and has been stable for over 60 months. The patient has not received any other treatment than surgery and oncolytic virotherapy. No severe adverse events have been reported during virotherapy. The present case suggests that ECHO-7 virotherapy is an effective treatment of skin melanoma.
Collapse
Affiliation(s)
| | | | - Evija Olmane
- Department of Radiology, Pauls Stradiņš Clinical University Hospital, Riga, Latvia
| | - Sergejs Isajevs
- Department of Pathology, Riga Eastern Clinical University Hospital, Riga, Latvia.,Department of Pathology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Tatjana Zablocka
- Department of Pathology, Riga Eastern Clinical University Hospital, Riga, Latvia.,Department of Pathology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | | | | |
Collapse
|
26
|
Efficacy and Safety of Oncolytic Viruses in Randomized Controlled Trials: A Systematic Review and Meta-Analysis. Cancers (Basel) 2020; 12:cancers12061416. [PMID: 32486227 PMCID: PMC7352817 DOI: 10.3390/cancers12061416] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic virotherapy is a promising antitumor therapeutic strategy. It is based on the ability of viruses to selectively kill cancer cells and induce host antitumor immune responses. However, the clinical outcomes of oncolytic viruses (OVs) vary widely. Therefore, we performed a meta-analysis to illustrate the efficacy and safety of oncolytic viruses. The Cochrane Library, PubMed, and EMBASE databases were searched for randomized controlled trials (RCTs) published up to 31 January 2020. The data for objective response rate (ORR), overall survival (OS), progression-free survival (PFS), and adverse events (AEs) were independently extracted by two investigators from 11 studies that met the inclusion criteria. In subgroup analyses, the objective response rate benefit was observed in patients treated with oncolytic DNA viruses (odds ratio (OR) = 4.05; 95% confidence interval (CI): 1.96–8.33; p = 0.0002), but not in those treated with oncolytic RNA viruses (OR = 1.00, 95% CI: 0.66–1.52, p = 0.99). Moreover, the intratumoral injection arm yielded a statistically significant improvement (OR = 4.05, 95% CI: 1.96–8.33, p = 0.0002), but no such improvement was observed for the intravenous injection arm (OR = 1.00, 95% CI: 0.66–1.52, p = 0.99). Among the five OVs investigated in RCTs, only talimogene laherparepvec (T-VEC) effectively prolonged the OS of patients (hazard ratio (HR), 0.79; 95% CI: 0.63–0.99; p = 0.04). None of the oncolytic virotherapies improved the PFS (HR = 1.00, 95% CI: 0.85–1.19, p = 0.96). Notably, the pooled rate of severe AEs (grade ≥3) was higher for the oncolytic virotherapy group (39%) compared with the control group (27%) (risk difference (RD), 12%; risk ratio (RR), 1.44; 95% CI: 1.17–1.78; p = 0.0006). This review offers a reference for fundamental research and clinical treatment of oncolytic viruses. Further randomized controlled trials are needed to verify these results.
Collapse
|
27
|
Abstract
OPINION STATEMENT There is an unmet need for additional treatments for metastatic melanoma, besides anti-PD1 antibodies which are FDA approved for adjuvant therapy for stage III or resected stage IV melanoma. Talimogene laherparepvec (T-VEC) is the first and only FDA-approved oncolytic virus for the treatment of melanoma. New viral vectors including coxsackieviruses, HF-10, adenovirus, reovirus, echovirus, and newcastle disease virus are currently under active development and investigation with varying degrees of efficacy in targeting melanoma. The use of T-VEC as a neoadjuvant therapy is emerging, but more data is needed at this point. T-VEC has also shown promise for use in combination therapy with ipilimumab, as T-VEC plus ipilimumab has a significantly higher objective response compared to ipilimumab alone. Data comparing T-VEC in combination with PD-1 checkpoint inhibitors is awaited, and a phase III trial is underway. It is likely that oncolytic viruses will have long-term application in the treatment of melanoma and that T-VEC in particular will continue to have a role in the treatment of patients with readily accessible cutaneous lesions both for local control and synergistic induction of antitumor immunity as part of combination therapies.
Collapse
Affiliation(s)
- Megan H Trager
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Larisa J Geskin
- Department of Dermatology, Columbia University, New York, NY, 10032, USA
| | - Yvonne M Saenger
- Department of Medicine, Division of Hematology Oncology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
28
|
Current Advances in the Treatment of BRAF-Mutant Melanoma. Cancers (Basel) 2020; 12:cancers12020482. [PMID: 32092958 PMCID: PMC7072236 DOI: 10.3390/cancers12020482] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/02/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most lethal form of skin cancer. Melanoma is usually curable with surgery if detected early, however, treatment options for patients with metastatic melanoma are limited and the five-year survival rate for metastatic melanoma had been 15-20% before the advent of immunotherapy. Treatment with immune checkpoint inhibitors has increased long-term survival outcomes in patients with advanced melanoma to as high as 50% although individual response can vary greatly. A mutation within the MAPK pathway leads to uncontrollable growth and ultimately develops into cancer. The most common driver mutation that leads to this characteristic overactivation in the MAPK pathway is the B-RAF mutation. Current combinations of BRAF and MEK inhibitors that have demonstrated improved patient outcomes include dabrafenib with trametinib, vemurafenib with cobimetinib or encorafenib with binimetinib. Treatment with BRAF and MEK inhibitors has met challenges as patient responses began to drop due to the development of resistance to these inhibitors which paved the way for development of immunotherapies and other small molecule inhibitor approaches to address this. Resistance to these inhibitors continues to push the need to expand our understanding of novel mechanisms of resistance associated with treatment therapies. This review focuses on the current landscape of how resistance occurs with the chronic use of BRAF and MEK inhibitors in BRAF-mutant melanoma and progress made in the fields of immunotherapies and other small molecules when used alone or in combination with BRAF and MEK inhibitors to delay or circumvent the onset of resistance for patients with stage III/IV BRAF mutant melanoma.
Collapse
|
29
|
Ammour Y, Ryabaya O, Shchetinina Y, Prokofeva E, Gavrilova M, Khochenkov D, Vorobyev D, Faizuloev E, Shohin I, Zverev VV, Svitich O, Nasedkina T. The Susceptibility of Human Melanoma Cells to Infection with the Leningrad-16 Vaccine Strain of Measles Virus. Viruses 2020; 12:E173. [PMID: 32033013 PMCID: PMC7077263 DOI: 10.3390/v12020173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/27/2020] [Accepted: 01/31/2020] [Indexed: 01/28/2023] Open
Abstract
Oncolytic viruses, including live attenuated measles virus (MV) vaccine strains, have recently been shown as promising therapeutic agents against human malignancies. In this study, the oncolytic potential of the attenuated vaccine strain Leningrad-16 (L-16) of MV was evaluated in a panel of human metastatic melanoma cell lines. The L-16 measles virus was shown to replicate within melanoma cells mediating direct cell killing of tumor cells, although all melanoma cell lines varied in regard to their ability to respond to L-16 MV infection, as revealed by the different pattern of the Interferon Stimulated Gene expression, cytokine release and mechanisms of cell death. Furthermore, the statistically significant L-16 measles virus related tumor growth inhibition was demonstrated in a melanoma xenograft model. Therefore, L-16 MV represents an appealing oncolytic platform for target delivery of therapeutic genes along with other attenuated measles virus strains.
Collapse
Affiliation(s)
- Yulia Ammour
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
| | - Oxana Ryabaya
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (O.R.); (D.K.)
| | - Yulia Shchetinina
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
| | - Elena Prokofeva
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
| | - Marina Gavrilova
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
| | - Dmitry Khochenkov
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (O.R.); (D.K.)
- Medicinal Chemistry Center, Togliatti State University, 445020 Togliatti, Russia
| | - Denis Vorobyev
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, 119146 Moscow, Russia
| | - Evgeny Faizuloev
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
| | - Igor Shohin
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
- Institute of biochemical technology and nanotechnology, RUDN University, 117198 Moscow, Russia
| | - Vitaly V. Zverev
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, 119146 Moscow, Russia
| | - Oxana Svitich
- I.I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia; (Y.S.); (E.P.); (M.G.); (D.V.); (E.F.); (I.S.); (V.V.Z.); (O.S.)
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, 119146 Moscow, Russia
| | - Tatiana Nasedkina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia;
| |
Collapse
|
30
|
Rahman MM, McFadden G. Oncolytic Virotherapy with Myxoma Virus. J Clin Med 2020; 9:jcm9010171. [PMID: 31936317 PMCID: PMC7020043 DOI: 10.3390/jcm9010171] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/25/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses are one of the most promising novel therapeutics for malignant cancers. They selectively infect and kill cancer cells while sparing the normal counterparts, expose cancer- specific antigens and activate the host immune system against both viral and tumor determinants. Oncolytic viruses can be used as monotherapy or combined with existing cancer therapies to become more potent. Among the many types of oncolytic viruses that have been developed thus far, members of poxviruses are the most promising candidates against diverse cancer types. This review summarizes recent advances that are made with oncolytic myxoma virus (MYXV), a member of the Leporipoxvirus genus. Unlike other oncolytic viruses, MYXV infects only rabbits in nature and causes no harm to humans or any other non-leporid animals. However, MYXV can selectively infect and kill cancer cells originating from human, mouse and other host species. This selective cancer tropism and safety profile have led to the testing of MYXV in various types of preclinical cancer models. The next stage will be successful GMP manufacturing and clinical trials that will bring MYXV from bench to bedside for the treatment of currently intractable malignancies.
Collapse
|
31
|
Sobhanimonfared F, Bamdad T, Sadigh ZA, Choobin H. Virus specific tolerance enhanced efficacy of cancer immuno-virotherapy. Microb Pathog 2019; 140:103957. [PMID: 31891795 DOI: 10.1016/j.micpath.2019.103957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 12/22/2019] [Accepted: 12/27/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Activation of the immune system to fight cancer is a major goal in immunology and oncology. Although cancer treatment using oncolytic viruses shows promising results, virus mediated oncolysis induces a weak anti-tumor immune response. Upon application of viruses, immune responses against the virus play a significant role in limiting tumor virotherapy. Although suppression of host immunity increases the efficacy of virotherapy against the tumor, but inhibits anti-tumor immune responses. Induction of viral specific tolerance before viral replication may cause the virus to efficiently replicate in tumor cells without affecting the immune responses against tumor antigens. Investigation of the combined strategy of virotherapy and immunotherapy using irradiated tumor cells along with IL-2 and interferon-alpha in virus specific tolerant mice was the goal of this study. MATERIALS AND METHODS For tolerance induction, the newborn mice were injected with vesicular stomatitis virus (VSV) subcutaneously. After injection of TC-1 tumor cells to adult tolerant mice and formation of a tumor, irradiated TC-1 cells along with IL-2 and Interferon-alpha expression plasmid were injected twice in mice and followed by virotherapy. Size of tumors and CTL activity against the virus and tumor cells were measured. RESULT The results showed increased efficacy of virotherapy in combination with immune-stimulators and tumor cells injection in tolerant mice compared to normal mice. CONCLUSION Specific tolerance against the oncolytic virus enhances the efficacy of virotherapy both in monotherapy and in combination with immunotherapy.
Collapse
Affiliation(s)
- Fatemeh Sobhanimonfared
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Taravat Bamdad
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Zohreh Azita Sadigh
- Human Viral Vaccine Department, Razi Vaccine and Serum Research Institute, Karaj, Iran.
| | - Hamzeh Choobin
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
32
|
How We Do It: Administration Guide for Intralesional Immunotherapy With Talimogene Laherparepvec (T-VEC) for Advanced Melanoma. Dermatol Surg 2019; 46:1455-1457. [PMID: 31403542 DOI: 10.1097/dss.0000000000002098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Kwiatkowska D, Kluska P, Reich A. Beyond PD-1 Immunotherapy in Malignant Melanoma. Dermatol Ther (Heidelb) 2019; 9:243-257. [PMID: 30927248 PMCID: PMC6522569 DOI: 10.1007/s13555-019-0292-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Indexed: 01/31/2023] Open
Abstract
For many years, the standard therapy for malignant melanoma was based mainly on surgical resection. Unfortunately, this treatment is curative only in the early localized stage of this malignancy. The metastatic stage of malignant melanoma still remains a huge therapeutic challenge. Despite the many new therapeutic options that have become available over the last years, there is a constant need for safer and more effective treatment modalities. There has been a dynamic development of various anti-cancer immunotherapies directed against new molecular targets. A number of clinical trials are currently being conducted to confirm their effectiveness and safety. In this review of the literature, we summarize the contemporary knowledge on promising new immunotherapies beyond the currently available treatment options for malignant melanoma, including oncolytic immunotherapy, selective inhibitors of indoleamine 2,3-dioxygenease, anti-PD-(L)1 (programmed death ligand 1) drugs, immune checkpoint protein LAG-3 antibodies, inhibitors of histone deacetylase (HDAC) and inhibitors of B7-H3.
Collapse
Affiliation(s)
| | - Piotr Kluska
- Wroclaw University of Science and Technology, Wrocław, Poland
| | - Adam Reich
- Department of Dermatology, University of Rzeszow, Rzeszów, Poland.
| |
Collapse
|