51
|
Tabolacci C, De Vita D, Facchiano A, Bozzuto G, Beninati S, Failla CM, Di Martile M, Lintas C, Mischiati C, Stringaro A, Del Bufalo D, Facchiano F. Phytochemicals as Immunomodulatory Agents in Melanoma. Int J Mol Sci 2023; 24:2657. [PMID: 36768978 PMCID: PMC9916941 DOI: 10.3390/ijms24032657] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Cutaneous melanoma is an immunogenic highly heterogenic tumor characterized by poor outcomes when it is diagnosed late. Therefore, immunotherapy in combination with other anti-proliferative approaches is among the most effective weapons to control its growth and metastatic dissemination. Recently, a large amount of published reports indicate the interest of researchers and clinicians about plant secondary metabolites as potentially useful therapeutic tools due to their lower presence of side effects coupled with their high potency and efficacy. Published evidence was reported in most cases through in vitro studies but also, with a growing body of evidence, through in vivo investigations. Our aim was, therefore, to review the published studies focused on the most interesting phytochemicals whose immunomodulatory activities and/or mechanisms of actions were demonstrated and applied to melanoma models.
Collapse
Affiliation(s)
- Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Daniela De Vita
- Department of Environmental Biology, University of Rome La Sapienza, 00185 Rome, Italy
| | | | - Giuseppina Bozzuto
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico, 00128 Rome, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, School of Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
52
|
Rao YJ, Goodman JF, Haroun F, Bauman JE. Integrating Immunotherapy into Multimodal Treatment of Head and Neck Cancer. Cancers (Basel) 2023; 15:cancers15030672. [PMID: 36765627 PMCID: PMC9913370 DOI: 10.3390/cancers15030672] [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: 12/01/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Patients with locally advanced head and neck squamous cell carcinoma (HNSCC) have a poor prognosis, with a significant risk of progression or death despite multimodal treatment with surgery, chemotherapy, and radiotherapy. Immune checkpoint inhibitors targeting the programmed death receptor-1 (PD1) have dramatically changed the treatment landscape for recurrent/metastatic disease, improving overall survival in both the first- and second-line palliative settings. This success has driven the investigation of treatment strategies incorporating immunotherapy earlier into the multimodal curative-intent or salvage treatment of both locally advanced and recurrent/metastatic HNSCC. This review encompassed the following three subjects, with a focus on recently reported and ongoing clinical trials: (1) the use of neoadjuvant immunotherapy prior to surgery for locally advanced HNSCC, (2) the use of immunochemoradiotherapy for locally advanced head and neck cancers, and (3) novel uses of immunotherapy in the salvage of recurrent/metastatic HNSCC via a combined modality, including reirradiation paradigms. The results of these studies are eagerly awaited to improve patient outcomes in this challenging disease.
Collapse
Affiliation(s)
- Yuan James Rao
- Division of Radiation Oncology, The George Washington University School of Medicine, Washington, DC 20037, USA
| | - Joseph F. Goodman
- Division of Head and Neck Surgery, The George Washington University School of Medicine, Washington, DC 20037, USA
| | - Faysal Haroun
- Division of Hematology/Oncology, The George Washington University School of Medicine, Washington, DC 20037, USA
| | - Julie E. Bauman
- Division of Hematology/Oncology, The George Washington University School of Medicine, Washington, DC 20037, USA
- Correspondence:
| |
Collapse
|
53
|
Schwartz GK. Flashback Foreword: Adjuvant Interferon α-2b for High-Risk Melanoma. J Clin Oncol 2023; 41:423-424. [PMID: 36649674 DOI: 10.1200/jco.22.02354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Gary K Schwartz
- Columbia University, New York, NY.,Associate Editor, Journal of Clinical Oncology, Alexandria, VA
| |
Collapse
|
54
|
Ma W, Xue R, Zhu Z, Farrukh H, Song W, Li T, Zheng L, Pan CX. Increasing cure rates of solid tumors by immune checkpoint inhibitors. Exp Hematol Oncol 2023; 12:10. [PMID: 36647169 PMCID: PMC9843946 DOI: 10.1186/s40164-023-00372-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
Immunotherapy has become the central pillar of cancer therapy. Immune checkpoint inhibitors (ICIs), a major category of tumor immunotherapy, reactivate preexisting anticancer immunity. Initially, ICIs were approved only for advanced and metastatic cancers in the salvage setting after or concurrent with chemotherapy at a response rate of around 20-30% with a few exceptions. With significant progress over the decade, advances in immunotherapy have led to numerous clinical trials investigating ICIs as neoadjuvant and/or adjuvant therapies for resectable solid tumors. The promising results of these trials have led to the United States Food and Drug Administration (FDA) approvals of ICIs as neoadjuvant or adjuvant therapies for non-small cell lung cancer, melanoma, triple-negative breast cancer, and bladder cancer, and the list continues to grow. This therapy represents a paradigm shift in cancer treatment, as many early-stage cancer patients could be cured with the introduction of immunotherapy in the early stages of cancer. Therefore, this topic became one of the main themes at the 2021 China Cancer Immunotherapy Workshop co-organized by the Chinese American Hematologist and Oncologist Network, the China National Medical Products Administration and the Tsinghua University School of Medicine. This review article summarizes the current landscape of ICI-based immunotherapy, emphasizing the new clinical developments of ICIs as curative neoadjuvant and adjuvant therapies for early-stage disease.
Collapse
Affiliation(s)
- Weijie Ma
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Ruobing Xue
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Ellis Fischel Cancer Center, University of Missouri, 1 Hospital Dr, Columbia, MO, 65201, USA
| | - Zheng Zhu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hizra Farrukh
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Kira Pharmaceuticals, Cambridge, MA, USA
| | - Tianhong Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Department of Medicine, Division of Hematology & Oncology, University of California Davis, Sacramento, CA, 95817, USA. .,Department of Medicine, VA Northern California Health Care System, Mather, CA, USA.
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,The Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Chong-Xian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,VA Boston Healthcare System, Boston, MA, 02132, USA. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
55
|
Ben Aïssa A. Immunotherapy in Melanoma: Highlights for the General Practitioner. PRAXIS 2023; 112:135-142. [PMID: 36855885 DOI: 10.1024/1661-8157/a003972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Melanoma is the most aggressive skin cancer, and surgery is the standard of care for localised disease. However, a risk of local and distant relapse exists despite tumour removal, particularly with thick or ulcerated tumours or lymph node involvement. Immunotherapy with immune checkpoint inhibitors (ICIs) targeting PD-1, PD-L-1 or CTLA-4 demonstrated improved relapse-free survival and distant metastasis-free survival against placebo after surgery for stage-III and high-risk stage-II melanoma. In unresectable localised and metastatic tumours, the double immunotherapy with ICIs (anti-PD-1+ anti-CTLA-4) allows for long-term survival in more than 50% of the patients. Novel immunotherapies (anti-LAG-3 ICI, adoptive cell therapy, intra-tumoural immunotherapy, cancer vaccines) and new combinations are in development to overcome resistance and improve patients' survival. Therapeutic decisions for each patient should be discussed in a specialised multidisciplinary team.
Collapse
Affiliation(s)
- Assma Ben Aïssa
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| |
Collapse
|
56
|
Sun J, Zhou JS, Wang YC, Dai HY, Sun MY, Lv C. Prognostic significance of age on superficial spreading melanoma after resection: lessons from SEER database involving 12 536 patients. ANZ J Surg 2023; 93:227-234. [PMID: 36368699 DOI: 10.1111/ans.18135] [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: 03/18/2022] [Revised: 08/15/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND This study sought to analyse the impact of elderly age on long-term prognosis of superficial spreading melanoma (SSM) after surgery. METHODS A population-based cohort of patients undergoing resection for SSM from 2004 to 2015 was collected, using data from National Cancer Institute' Surveillance, Epidemiology, and End Results (SEER)* Stat software. Patients were divided into the non-elderly group (≤70 years) and elderly group (>70 years). Baseline characteristics and long-term survivals were compared between the two groups. A 1:1 propensity score matching (PSM) was used to reduce the risk of bias. The impact of the elderly age on overall survival (OS) and cause-specific mortality (CSM) was estimated by Cox-regression and competing-risk regression models. RESULTS Among 12 536 patients with SSM after resection included into the cohort, 8664 patients were ≤70 years, and 3872 were >70 years. Patients in the elderly group had higher incidences of multiple tumours, worse tumour stage and infiltration degree, lymphatic metastasis, and larger size of primary lesions. Using PSM, 3581 pairs of patients were created. On matched analysis, the elderly group was associated with worse OS and CSM. On multivariable Cox-regression and competing-risk regression analyses, elderly age was identified as an independent risk factor of OS and CSM after adjusting for other prognostic variables. CONCLUSIONS The elderly age of patients was independently associated with worse OS and CSM after resection of SSM when baseline and tumour characteristics were balanced. Adjuvant therapy and individualized strategy on follow-up should be made for elderly patients after resection of SSM.
Collapse
Affiliation(s)
- Jie Sun
- Department of Plastic Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Jie-Song Zhou
- Department of Plastic Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Yu-Chong Wang
- Department of Plastic Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Hai-Ying Dai
- Department of Plastic Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Meng-Yan Sun
- Department of Plastic Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Chuan Lv
- Department of Plastic Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| |
Collapse
|
57
|
Impact of Precision Medicine in Oncology: Immuno-oncology. Cancer J 2023; 29:15-19. [PMID: 36693153 DOI: 10.1097/ppo.0000000000000641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
ABSTRACT Cancer treatment has dramatically changed over the last decade with the development of immunotherapy. Therapies including immune cytokines, immune checkpoint inhibition, intratumoral therapies, and cellular therapies are already widely used in the oncology clinic. Active development continues in these areas and in the development of vaccines, bispecific therapies, and more refined cellular therapies. In this review, we will examine the role that immune therapy has in cancer treatment and explore areas of future development.
Collapse
|
58
|
McLouth LE, Zheng Y, Smith S, Hodi FS, Rao UN, Cohen GI, Amatruda TT, Dakhil SR, Curti BD, Nakhoul I, Chandana SR, Bane CL, Marinier DE, Lee SJ, Sondak VK, Kirkwood JM, Tarhini AA, Wagner LI. Patient-reported tolerability of adjuvant ipilimumab (3 or 10 mg/kg) versus high-dose interferon alfa-2b for resected high-risk stage III-IV melanoma in phase III trial E1609. Qual Life Res 2023; 32:183-196. [PMID: 36029412 PMCID: PMC9839512 DOI: 10.1007/s11136-022-03226-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 01/17/2023]
Abstract
PURPOSE Trial E1609 demonstrated superior overall survival with ipilimumab 3 mg/kg (ipi3) compared to high-dose interferon (HDI) for patients with resected high-risk melanoma. To inform treatment tolerability, we compared health-related quality of life (HRQoL), gastrointestinal (GI), and treatment-specific physical and cognitive/emotional symptoms. We also compared treatment-specific concerns between all arms. METHODS We assessed HRQoL using the Functional Assessment of Cancer Therapy-General, physical and cognitive/emotional concerns using the FACT-Biologic Response Modifier subscale, and GI symptoms with the Functional Assessment of Chronic Illness Therapy-Diarrhea subscale pre-treatment and every 3 months. The primary outcome was the difference in HRQoL at 3 months between ipi3/ipi10 vs. HDI. RESULTS 549 patients (n = 158 ipi3; n = 191 ipi10; n = 200 HDI) were analyzed. 3-month completion was 58.7%. Compared to HDI, ipilimumab patients reported better HRQoL (ipi3 = 87.5 ± 14.6 vs. HDI = 74.7 ± 15.4, p < .001; ipi10 = 84.9 ± 16.5 vs. HDI, p < .001) and fewer physical (ipi3 = 22.3 ± 4.6 vs. HDI = 17.1 ± 5.4, p < .001; ipi10 = 21.8 ± 5.0 vs. HDI p < .001) and cognitive/emotional (ipi3 = 18.6 ± 4.4 vs. HDI = 15.0 ± 5.3, p < .001; ipi10 = 17.7 ± 4.8 vs. HDI p < .001) concerns, but worse GI symptoms (ipi3 = 40.8 ± 5.0 vs. HDI = 42.2 ± 2.9, p = .011; ipi10 = 39.5 ± 7.0 vs. HDI, p < .001). Fewer ipilimumab patients reported worsening treatment-specific concerns (e.g., 52% of ipi3 and 58% of ipi10 reported worsening fatigue vs. 82% HDI, p's < .001). CONCLUSION PROs demonstrated less toxicity of ipi3 compared to HDI and ipi10. Priorities for symptom management among patients receiving ipilimumab include GI toxicities, fatigue, weakness, appetite loss, arthralgia, and depression. TRIAL REGISTRATION NCT01274338, January 11, 2011 (first posted date) https://clinicaltrials.gov/ct2/show/NCT01274338?term=NCT01274338&draw=2&rank=1 .
Collapse
Affiliation(s)
- Laurie E McLouth
- Department of Behavioral Science, College of Medicine, Markey Cancer Center, University of Kentucky, 467 Healthy Kentucky Research Building, 760 Press Avenue, Lexington, KY, 40508, USA.
| | - Yue Zheng
- Dana-Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA, USA
| | - Stephanie Smith
- Nancy N. and J.C. Lewis Cancer and Research Pavilion, St. Joseph's/Candler, Savannah, GA, USA
| | - F Stephen Hodi
- Dana-Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA, USA
- Dana-Farber Cancer Institute/Harvard Cancer Center, Boston, MA, USA
| | - Uma N Rao
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Gary I Cohen
- Greater Baltimore Medical Center, Baltimore, MD, USA
| | | | | | - Brendan D Curti
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Ibrahim Nakhoul
- Regional Cancer Center at Indian Path Community Hospital, Kingsport, TN, USA
| | - Sreenivasa R Chandana
- Cancer and Hematology Centers of Western Michigan/Cancer Research Consortium of West Michigan NCORP, Grand Rapids, MI, USA
| | | | | | - Sandra J Lee
- Dana-Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA, USA
| | | | - John M Kirkwood
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | - Lynne I Wagner
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| |
Collapse
|
59
|
Kudling TV, Clubb JH, Quixabeira DC, Santos JM, Havunen R, Kononov A, Heiniö C, Cervera-Carrascon V, Pakola S, Basnet S, Grönberg-Vähä-Koskela S, Arias V, Gladwyn-Ng I, Aro K, Bäck L, Räsänen J, Ilonen I, Borenius K, Räsänen M, Hemminki O, Rannikko A, Kanerva A, Tapper J, Hemminki A. Local delivery of interleukin 7 with an oncolytic adenovirus activates tumor-infiltrating lymphocytes and causes tumor regression. Oncoimmunology 2022; 11:2096572. [PMID: 35845722 PMCID: PMC9278414 DOI: 10.1080/2162402x.2022.2096572] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cytokines have proven to be effective for cancer therapy, however whilst low-dose monotherapy with cytokines provides limited therapeutic benefit, high-dose treatment can lead to a number of adverse events. Interleukin 7 has shown promising results in clinical trials, but anti-cancer effect was limited, in part due to a low concentration of the cytokine within the tumor. We hypothesized that arming an oncolytic adenovirus with Interleukin 7, enabling high expression localized to the tumor microenvironment, would overcome systemic delivery issues and improve therapeutic efficacy. We evaluated the effects of Ad5/3-E2F-d24-hIL7 (TILT-517) on tumor growth, immune cell activation and cytokine profiles in the tumor microenvironment using three clinically relevant animal models and ex vivo tumor cultures. Our data showed that local treatment of tumor bearing animals with Ad5/3- E2F-d24-hIL7 significantly decreased cancer growth and increased frequency of tumor-infiltrating cells. Ad5/3-E2F-d24-hIL7 promoted notable upregulation of pro-inflammatory cytokines, and concomitant activation and migration of CD4+ and CD8 + T cells. Interleukin 7 expression within the tumor was positively correlated with increased number of cytotoxic CD4+ cells and IFNg-producing CD4+ and CD8+ cells. These findings offer an approach to overcome the current limitations of conventional IL7 therapy and could therefore be translated to the clinic.
Collapse
Affiliation(s)
- Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - James H.A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Dafne C.A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Alexander Kononov
- Systems Oncology Group, Cancer research UK, Manchester University, Manchester, UK
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Katri Aro
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Otorhinolaryngology – Head and Neck Surgery, Helsinki Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Leif Bäck
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Otorhinolaryngology – Head and Neck Surgery, Helsinki Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jari Räsänen
- Department of Thoracic Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilkka Ilonen
- Department of Thoracic Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristian Borenius
- Department of Thoracic Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko Räsänen
- Department of Thoracic Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Otto Hemminki
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Antti Rannikko
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Johanna Tapper
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| |
Collapse
|
60
|
Advances of Electroporation-Related Therapies and the Synergy with Immunotherapy in Cancer Treatment. Vaccines (Basel) 2022; 10:vaccines10111942. [PMID: 36423037 PMCID: PMC9692484 DOI: 10.3390/vaccines10111942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Electroporation is the process of instantaneously increasing the permeability of a cell membrane under a pulsed electric field. Depending on the parameters of the electric pulses and the target cell electrophysiological characteristics, electroporation can be either reversible or irreversible. Reversible electroporation facilitates the delivery of functional genetic materials or drugs to target cells, inducing cell death by apoptosis, mitotic catastrophe, or pseudoapoptosis; irreversible electroporation is an ablative technology which directly ablates a large amount of tissue without causing harmful thermal effects; electrotherapy using an electric field can induce cell apoptosis without any aggressive invasion. Reversible and irreversible electroporation can also activate systemic antitumor immune response and enhance the efficacy of immunotherapy. In this review, we discuss recent progress related to electroporation, and summarize its latest applications. Further, we discuss the synergistic effects of electroporation-related therapies and immunotherapy. We also propose perspectives for further investigating electroporation and immunotherapy in cancer treatment.
Collapse
|
61
|
Su CL, Chiou SH, Lin FC, Platt RW. Analysis of survival data with cure fraction and variable selection: A pseudo-observations approach. Stat Methods Med Res 2022; 31:2037-2053. [PMID: 35754373 PMCID: PMC9660265 DOI: 10.1177/09622802221108579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
In biomedical studies, survival data with a cure fraction (the proportion of subjects cured of disease) are commonly encountered. The mixture cure and bounded cumulative hazard models are two main types of cure fraction models when analyzing survival data with long-term survivors. In this article, in the framework of the Cox proportional hazards mixture cure model and bounded cumulative hazard model, we propose several estimators utilizing pseudo-observations to assess the effects of covariates on the cure rate and the risk of having the event of interest for survival data with a cure fraction. A variable selection procedure is also presented based on the pseudo-observations using penalized generalized estimating equations for proportional hazards mixture cure and bounded cumulative hazard models. Extensive simulation studies are conducted to examine the proposed methods. The proposed technique is demonstrated through applications to a melanoma study and a dental data set with high-dimensional covariates.
Collapse
Affiliation(s)
- Chien-Lin Su
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish
General Hospital, Montréal, Québec, Canada
- Peri and Post Approval Studies, Strategic and Scientific Affairs,
PPD, part of Thermo Fisher Scientific, Montréal, Québec, Canada
| | - Sy Han Chiou
- Department of Mathematical Sciences, University of Texas at Dallas,
Richardson, TX, USA
| | - Feng-Chang Lin
- Department of Biostatistics, University of North Carolina, Chapel
Hill, NC, USA
| | - Robert W Platt
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish
General Hospital, Montréal, Québec, Canada
| |
Collapse
|
62
|
Kobeissi I, Tarhini AA. Systemic adjuvant therapy for high-risk cutaneous melanoma. Ther Adv Med Oncol 2022; 14:17588359221134087. [PMID: 36324735 PMCID: PMC9619267 DOI: 10.1177/17588359221134087] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Cutaneous melanoma continues to increase in incidence and poses a significant mortality risk. Surgical excision of melanoma in its early stages is often curative. However, patients with resected stages IIB-IV are considered at high risk for relapse and death from melanoma where systemic adjuvant therapy is indicated. The long-studied high-dose interferon-α was shown to improve relapse-free survival (RFS) and overall survival (OS) but is no longer in use. Adjuvant therapy with ipilimumab at 10 mg/kg (ipi10) demonstrated significant RFS and OS improvements but at a high cost in terms of toxicity, while adjuvant ipilimumab 3 mg/kg was shown to be equally effective and less toxic. More recently, the adjuvant therapy for resected stages III-IV melanoma in clinical practice has changed in favor of nivolumab, pembrolizumab, and BRAF-MEK inhibitors dabrafenib plus trametinib (for BRAF mutant melanoma) based on significant improvements in RFS as compared to ipi10 (nivolumab and pembrolizumab) and placebo (dabrafenib plus trametinib). For resected stages IIB-IIC melanoma, pembrolizumab achieved regulatory approval in the United States based on significant RFS benefits. In this article, we review completed and ongoing phase III adjuvant therapy trials. We also briefly discuss neoadjuvant therapy for locoregionally advanced melanoma. Finally, we explore recent studies on predictive and prognostic melanoma biomarkers in the adjuvant setting.
Collapse
Affiliation(s)
- Iyad Kobeissi
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | |
Collapse
|
63
|
Early Effects of Nivolumab and Ipilimumab Combined Immunotherapy in the Treatment of Metastatic Melanoma in Poland: A Multicenter Experience. Biomedicines 2022; 10:biomedicines10102528. [PMID: 36289790 PMCID: PMC9598919 DOI: 10.3390/biomedicines10102528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Nivolumab and ipilimumab combination became the first-line standard in advanced melanoma. We assessed its efficacy in a real-life study in Poland. In a one-year follow-up, we evaluated the medical records of 50 melanoma patients treated with that modality in five oncology centers. We recorded therapy outcomes and adverse events (AEs) after 3 and 12 months of therapy. At the first checkpoint, the disease control rate (DCR) was recorded in 58% (n = 29) of patients, but the same number of patients (n = 29, 58%) stopped immunotherapy due to disease progression (PD, n = 14, 48.3%), toxicity (n = 11, 37.9%) or death (n = 4, 13.8%). Among patients with DCR after the induction phase, 8 (27.6%) terminated due to toxicity, and 21 (72.4%) continued. However, at the 12-month checkpoint, only 14 patients (27% of all) were still receiving immunotherapy. In 7 (33.3%) it was discontinued due to PD (n = 2), toxicity (n = 2, 28.6% each), or death (n = 3, 42.9%). AEs occurred in 66.7% (n = 34) of patients; severe (grade 3 or 4) in half of them. Interestingly, those with AEs had an 80% lower risk of death (hazard ratio [HR] 0.2, 95% confidence interval [CI] 0.07−0.57, p = 0.001) and PD (HR 0.2, 95%CI 0.09−0.47, p < 0.0001). In the entire group of patients, after a 12-month follow-up, the median overall survival was not reached (NR, range: 6.8 months-NR) and progression-free survival was 6.3 (range: 3-NR) months. Our results demonstrate that combined immunotherapy is less effective in real-life than in pivotal trials. However, early responders will likely continue the therapy after a one-year follow-up. AEs occurrence might be a predictor of clinical effectiveness.
Collapse
|
64
|
Zhong W, Diao G. Semiparametric Density Ratio Model for Survival Data with a Cure Fraction. STATISTICS IN BIOSCIENCES 2022. [DOI: 10.1007/s12561-022-09357-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
65
|
Xiong F, Wang Q, Wu GH, Liu WZ, Wang B, Chen YJ. Direct and indirect effects of IFN-α2b in malignancy treatment: not only an archer but also an arrow. Biomark Res 2022; 10:69. [PMID: 36104718 PMCID: PMC9472737 DOI: 10.1186/s40364-022-00415-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Interferon-α2b (IFN-α2b) is a highly active cytokine that belongs to the interferon-α (IFN-α) family. IFN-α2b has beneficial antiviral, antitumour, antiparasitic and immunomodulatory activities. Direct and indirect antiproliferative effects of IFN-α2b have been found to occur via multiple pathways, mainly the JAK-STAT pathway, in certain cancers. This article reviews mechanistic studies and clinical trials on IFN-α2b. Potential regulators of the function of IFN-α2b were also reviewed, which could be utilized to relieve the poor response to IFN-α2b. IFN-α2b can function not only by enhancing the systematic immune response but also by directly killing tumour cells. Different parts of JAK-STAT pathway activated by IFN-α2b, such as interferon alpha and beta receptors (IFNARs), Janus kinases (JAKs) and IFN‐stimulated gene factor 3 (ISGF3), might serve as potential target for enhancing the pharmacological action of IFN-α2b. Despite some issues that remain to be solved, based on current evidence, IFN-α2b can inhibit disease progression and improve the survival of patients with certain types of malignant tumours. More efforts should be made to address potential adverse effects and complications.
Collapse
|
66
|
Chemotherapy in combination with anti-PD-1 agents as adjuvant therapy for high-risk oral mucosal melanoma. J Cancer Res Clin Oncol 2022; 149:2293-2300. [PMID: 36088610 DOI: 10.1007/s00432-022-04090-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/24/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Adjuvant therapy plays a critical role in the treatment of oral mucosal melanoma (OMM). Anti-programmed cell death-1 (PD-1) agents are recommended as front-line therapy for metastatic melanoma, but their efficacy as adjuvant therapy for high-risk OMM remains unclear. PATIENTS AND METHODS A single-center, retrospective cohort study was conducted in 193 nodular-type oral mucosal melanoma (NOMM) patients who received chemotherapy alone or in combination with high-dose interferon-α2b (HDI) or anti-PD-1 agents as adjuvant therapy. Multivariate analysis was performed to identify significant prognostic factors for the 2-year overall survival (OS) and progression-free survival (PFS). RESULTS Tumor thickness, ulceration and invasion level were found to be independent prognostic factors for both 2-year OS and PFS, while T-stage was only associated with OS. The 2-year OS and PFS were 43.5% and 10.9% in patients who received only chemotherapy. In comparison, the 2-year OS was improved, albeit not significantly (47.4%; p > 0.05), and PFS was significantly improved (43.6%; p = 0.0028) in patients who received chemotherapy plus HDI; and both 2-year OS (71.0%; p = 0.0118) and PFS (53.6%; p = 0.0001) were significantly improved in patients received chemotherapy plus anti-PD-1. The serious adverse event (SAE) (p < 0.0001) and discontinued treatment due to SAE (p < 0.0001) were significantly lower in patients who received anti-PD-1 than in patients who received HDI. CONCLUSIONS Invasion level and tumor thickness are independent prognostic factors for NOMM. Chemotherapy plus anti-PD-1 agents seem to be the adjuvant therapy of choice for NOMM, as it is safer and more tolerable than HDI and, more importantly, it can significantly improve the OS and PFS.
Collapse
|
67
|
Borówka M, Łącki-Zynzeling S, Nicze M, Kozak S, Chudek J. Adverse Renal Effects of Anticancer Immunotherapy: A Review. Cancers (Basel) 2022; 14:4086. [PMID: 36077623 PMCID: PMC9454552 DOI: 10.3390/cancers14174086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Modern oncological therapy utilizes various types of immunotherapy. Immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cells (CAR-T) therapy, cancer vaccines, tumor-targeting monoclonal antibodies (TT-mAbs), bispecific antibodies and cytokine therapy improve patients' outcomes. However, stimulation of the immune system, beneficial in terms of fighting against cancer, generates the risk of harm to other cells in a patient's body. Kidney damage belongs to the relatively rare adverse events (AEs). Best described, but still, superficially, are renal AEs in patients treated with ICIs. International guidelines issued by the European Society for Medical Oncology (ESMO) and the American Society of Clinical Oncology (ASCO) cover the management of immune-related adverse events (irAEs) during ICI therapy. There are fewer data concerning real occurrence and possible presentations of renal adverse drug reactions of other immunotherapeutic methods. This implies the need for the collection of safety data during ongoing clinical trials and in the real-life world to characterize the hazard related to the use of new immunotherapies and management of irAEs.
Collapse
Affiliation(s)
| | - Stanisław Łącki-Zynzeling
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Reymonta 8, 40-027 Katowice, Poland
| | | | | | | |
Collapse
|
68
|
Jung S, Johnson DB. Management of Acral and Mucosal Melanoma: Medical Oncology Perspective. Oncologist 2022; 27:703-710. [PMID: 35640549 PMCID: PMC9355814 DOI: 10.1093/oncolo/oyac091] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Acral and mucosal melanomas (MM) are rare subtypes of melanoma that are biologically and clinically distinct from cutaneous melanoma. Despite the progress in the treatment of cutaneous melanomas with the development of targeted and immune therapies, the therapeutic options for these less common subtypes remain limited. Difficulties in early diagnosis, the aggressive nature of the disease, and the frequently occult sites of origin have also contributed to the poor prognosis associated with acral and MM, with substantially worse long-term prognosis. The rarity of these subtypes has posed significant barriers to better understanding their biological features and investigating novel therapies. Consequently, establishing standardized treatment guidelines has been a challenge. In this review, we provide a brief overview of the current knowledge regarding acral and MM, focusing on their epidemiology, genetic backgrounds, and unique clinical characteristics. Further discussion centers around the management of primary and advanced disease and the role of emerging targeted and immune therapies for these subtypes, specifically focusing on issues relevant to medical oncologists.
Collapse
Affiliation(s)
- Seungyeon Jung
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville TN, USA
| |
Collapse
|
69
|
Lin EPY, Hsu CY, Berry L, Bunn P, Shyr Y. Analysis of Cancer Survival Associated With Immune Checkpoint Inhibitors After Statistical Adjustment: A Systematic Review and Meta-analyses. JAMA Netw Open 2022; 5:e2227211. [PMID: 35976648 PMCID: PMC9386543 DOI: 10.1001/jamanetworkopen.2022.27211] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
IMPORTANCE Appropriate clinical decision-making relies on accurate data interpretation, which in turn relies on the use of suitable statistical models. Long tails and early crossover-2 features commonly observed in immune checkpoint inhibitor (ICI) survival curves-raise questions as to the suitability of Cox proportional hazards regression for ICI survival analysis. Cox proportional hazards-Taylor expansion adjustment for long-term survival data (Cox-TEL) adjustment may provide possible solutions in this setting. OBJECTIVE To estimate overall survival and progression-free survival benefits of ICI therapy vs chemotherapy using Cox-TEL adjustment. DATA SOURCES A PubMed search was performed for all cataloged publications through May 22, 2022. STUDY SELECTION The search was restricted to randomized clinical trials with search terms for ICIs and lung cancer, melanoma, or urothelial carcinoma. The publications identified were further reviewed for inclusion. DATA EXTRACTION AND SYNTHESIS Cox proportional hazards ratios (HRs) were transformed to Cox-TEL HRs for patients with short-term treatment response (ie, short-term survivor) (ST-HR) and difference in proportions for patients with long-term survival (LT-DP) by Cox-TEL. Meta-analyses were performed using a frequentist random-effects model. MAIN OUTCOMES AND MEASURES Outcomes of interest were pooled overall survival (primary outcome) and progression-free survival (secondary outcome) HRs, ST-HRs, and LT-DPs. Subgroup analyses stratified by cancer type also were performed. RESULTS A total of 1036 publications was identified. After 3 levels of review against inclusion criteria, 13 clinical trials (7 in non-small cell lung cancer, 3 in melanoma, and 3 in urothelial carcinoma) were selected for the meta-analysis. In the primary analysis, pooled findings were 0.75 (95% CI, 0.70-0.81) for HR, 0.86 (95% CI, 0.81-0.92) for ST-HR, and 0.08 (95% CI, 0.06-0.10) for LT-DP. In the secondary analysis, the pooled values for progression-free survival were 0.77 (95% CI, 0.64-0.91) for HR, 1.02 (95% CI, 0.84-1.24) for ST-HR, and 0.10 (95% CI, 0.06-0.14) for LT-DP. CONCLUSIONS AND RELEVANCE This systematic review and meta-analysis of ICI clinical trial results noted consistently larger ST-HRs vs Cox HRs for ICI therapy, with an LT-DP of approximately 10%. These results suggest that Cox HRs may not provide a full picture of survival outcomes when the risk reduction from treatment is not constant, which may aid in the decision-making process of oncologists and patients.
Collapse
Affiliation(s)
- Emily Pei-Ying Lin
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Yuan Hsu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne Berry
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul Bunn
- Department of Medicine, University of Colorado School of Medicine, Aurora
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
70
|
Cytokine chemokine network in tumor microenvironment: Impact on CSC properties and therapeutic applications. Cytokine 2022; 156:155916. [DOI: 10.1016/j.cyto.2022.155916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/27/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022]
|
71
|
Sharon CE, Karakousis GC. Educational Review: Neoadjuvant Approaches to Melanoma. Ann Surg Oncol 2022; 29:8492-8500. [PMID: 35849287 DOI: 10.1245/s10434-022-12224-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/02/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND With the development of novel systemic therapies, the treatment of patients with melanoma has changed drastically over the past few years, especially with regard to neoadjuvant treatments. Standard of care for patients with resectable stage III/IV melanoma traditionally consisted of surgery, with possible adjuvant treatment. However, there have been promising improvements in patient outcomes with neoadjuvant treatment compared to upfront surgery, specifically with targeted and immune therapies. METHODS A review of clinical trials in the neoadjuvant treatment of stage III/IV melanoma was performed. RESULTS Multiple phase I-II clinical trials have investigated the utility of interferon, targeted therapies (i.e., BRAF and/or MEK inhibitors) and immune checkpoint inhibitors (i.e., PD-1 or CTLA-4 inhibitors) in the treatment of resectable clinical stage III/IV melanoma. Large strides have been made with regards to optimal treatment strategy and dosing, to maximize clinical and pathologic response rates while minimizing toxicities. Additionally, complete pathologic response to neoadjuvant therapies translates to a disease-free survival benefit. Current and future directions include individualizing surgical and adjuvant therapy based on patient response to neoadjuvant treatments. CONCLUSIONS The current evidence, represented by small phase I-II trials, demonstrates advantages to neoadjuvant treatment with targeted or immune therapy for patients with resectable stage III/IV melanoma. Future research is needed to determine the advantages of neoadjuvant compared to adjuvant treatment, and to further refine treatment strategies based on patient response.
Collapse
Affiliation(s)
- Cimarron E Sharon
- Division of Endocrine and Oncologic Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, Maloney 4, Philadelphia, PA, USA.
| | - Giorgos C Karakousis
- Division of Endocrine and Oncologic Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, Maloney 4, Philadelphia, PA, USA
| |
Collapse
|
72
|
Garbe C, Amaral T, Peris K, Hauschild A, Arenberger P, Basset-Seguin N, Bastholt L, Bataille V, Del Marmol V, Dréno B, Fargnoli MC, Forsea AM, Grob JJ, Hoeller C, Kaufmann R, Kelleners-Smeets N, Lallas A, Lebbé C, Lytvynenko B, Malvehy J, Moreno-Ramirez D, Nathan P, Pellacani G, Saiag P, Stratigos AJ, Van Akkooi ACJ, Vieira R, Zalaudek I, Lorigan P. European consensus-based interdisciplinary guideline for melanoma. Part 2: Treatment - Update 2022. Eur J Cancer 2022; 170:256-284. [PMID: 35623961 DOI: 10.1016/j.ejca.2022.04.018] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A unique collaboration of multidisciplinary experts from the European Dermatology Forum (EDF), the European Association of Dermato-Oncology (EADO), and the European Organization of Research and Treatment of Cancer (EORTC) was formed to make recommendations on cutaneous melanoma diagnosis and treatment, based on the systematic literature reviews and the experts' experience. Cutaneous melanomas are excised with one to 2-cm safety margins. Sentinel lymph node dissection shall be performed as a staging procedure in patients with tumor thickness ≥1.0 mm or ≥0.8 mm with additional histological risk factors, although there is as yet no clear survival benefit for this approach. Therapeutic decisions in stage III/IV patients should be primarily made by an interdisciplinary oncology team ("tumor board"). Adjuvant therapies can be proposed in stage III/completely resected stage IV patients and are primarily anti-PD-1, independent of mutational status, or alternatively dabrafenib plus trametinib for BRAF mutant patients. In distant metastases (stage IV), either resected or not, systemic treatment is always indicated. For first-line treatment particularly in BRAF wild-type patients, immunotherapy with PD-1 antibodies alone or in combination with CTLA-4 antibodies shall be considered. In stage IV melanoma with a BRAF-V600 E/K mutation, first-line therapy with BRAF/MEK inhibitors can be offered as an alternative to immunotherapy. In patients with primary resistance to immunotherapy and harboring a BRAF-V600 E/K mutation, this therapy shall be offered as second-line therapy. Systemic therapy in stage III/IV melanoma is a rapidly changing landscape, and it is likely that these recommendations may change in the near future.
Collapse
Affiliation(s)
- Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany.
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Ketty Peris
- Institute of Dermatology, Università Cattolica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Petr Arenberger
- Department of Dermatovenereology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicole Basset-Seguin
- Université Paris Cite, AP-HP, Department of Dermatology INSERM U 976 Hôpital, Saint Louis Paris France
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Veronique Bataille
- Twin Research and Genetic Epidemiology Unit, School of Basic & Medical Biosciences, King's College London, London, SE1 7EH, United Kingdom
| | - Veronique Del Marmol
- Department of Dermatology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Brigitte Dréno
- Dermatology Department, CHU Nantes, CIC 1413, CRCINA, University Nantes, Nantes, France
| | - Maria C Fargnoli
- Dermatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Ana-Maria Forsea
- Dermatology Department, Elias University Hospital, Carol Davila University of Medicine and Pharmacy Bucharest, Romania
| | | | | | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology, Frankfurt University Hospital, Frankfurt, Germany
| | | | - Aimilios Lallas
- First Department of Dermatology, Aristotle University, Thessaloniki, Greece
| | - Celeste Lebbé
- Université Paris Cite, AP-HP, Department of Dermatology INSERM U 976 Hôpital, Saint Louis Paris France
| | - Bodhan Lytvynenko
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Josep Malvehy
- Melanoma Unit, Department of Dermatology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - David Moreno-Ramirez
- Medical-&-Surgical Dermatology Service, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Paul Nathan
- Mount-Vernon Cancer Centre, Northwood United Kingdom
| | | | - Philippe Saiag
- University Department of Dermatology, Université de Versailles-Saint Quentin en Yvelines, APHP, Boulogne, France
| | - Alexander J Stratigos
- First Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Alexander C J Van Akkooi
- Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Ricardo Vieira
- Department of Dermatology and Venereology, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Iris Zalaudek
- Dermatology Clinic, Maggiore Hospital, University of Trieste, Trieste, Italy
| | - Paul Lorigan
- The University of Manchester, Oxford Rd, Manchester, M13 9PL, United Kingdom
| |
Collapse
|
73
|
Immunotherapy: an alternative promising therapeutic approach against cancers. Mol Biol Rep 2022; 49:9903-9913. [PMID: 35759082 PMCID: PMC9244230 DOI: 10.1007/s11033-022-07525-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/26/2022] [Indexed: 10/26/2022]
Abstract
The immune system interacts with cancer cells in multiple intricate ways that can shield the host against hyper-proliferation but can also contribute to malignancy. Understanding the protective roles of the immune system in its interaction with cancer cells can help device new and alternate therapeutic strategies. Many immunotherapeutic methodologies, including adaptive cancer therapy, cancer peptide vaccines, monoclonal antibodies, and immune checkpoint treatment, have transformed the traditional cancer treatment landscape. However, many questions remain unaddressed. The development of personalized combination therapy and neoantigen-based cancer vaccines would be the avant-garde approach to cancer treatment. Desirable chemotherapy should be durable, safe, and target-specific. Managing both tumor (intrinsic factors) and its microenvironment (extrinsic factors) are critical for successful immunotherapy. This review describes current approaches and their advancement related to monoclonal antibody-related clinical trials, new cytokine therapy, a checkpoint inhibitor, adoptive T cell therapy, cancer vaccine, and oncolytic virus.
Collapse
|
74
|
Lao CD, Khushalani NI, Angeles C, Petrella TM. Current State of Adjuvant Therapy for Melanoma: Less Is More, or More Is Better? Am Soc Clin Oncol Educ Book 2022; 42:1-7. [PMID: 35658502 DOI: 10.1200/edbk_351153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advances in melanoma treatments over the past decade have changed the course of survival for patients. Several adjuvant therapies have been approved and are now considered standard of care for high-risk patients. These therapies have shown improvements for recurrence-free survival and distant metastases-free survival, but not overall survival, as the data are maturing. The 5-year recurrence-free survival in the COMBI-AD study, which compared dabrafenib and trametinib with placebo, was 65% and 58%, respectively. In the KEYNOTE-054 study, the recurrence-free survival at 3 years was 63.7% versus 41%. Despite these advances, approximately 50% of patients will succumb to their disease. Adjuvant therapy is considered potentially curative and avoids the morbidity of relapsed disease and the poor outcomes seen in metastatic disease. However, the lack of overall survival benefit in clinical trials of patients with high-risk stage II and stage III disease raises the question of whether it is more efficacious to treat when there is residual microscopic disease, or to wait until the disease recurs to avoid treating those who may have been cured by surgery alone. Immunotherapy also has the potential for substantial toxicity that may be lifelong; hence, discussion of risks and benefits of therapy is warranted because there should be less tolerance for substantial toxicity in the adjuvant setting. Adjuvant trials are needed that will integrate biomarkers to allow for better selection of patients who will truly benefit from adjuvant therapy.
Collapse
Affiliation(s)
| | | | | | - Teresa M Petrella
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
75
|
Qureischi M, Mohr J, Arellano-Viera E, Knudsen SE, Vohidov F, Garitano-Trojaola A. mRNA-based therapies: Preclinical and clinical applications. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:1-54. [PMID: 36064262 DOI: 10.1016/bs.ircmb.2022.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
At the fundamental level, messenger RNA (mRNA)-based therapeutics involves the delivery of in vitro-transcribed (IVT) mRNA into the cytoplasm of a target cell, where it is translated into the desired protein. IVT mRNA presents various advantages compared to DNA and recombinant protein-based approaches that make it ideal for a broad range of therapeutic applications. IVT mRNA, which is translated in the cytoplasm after transfection into cells, can encode virtually any target protein. Notably, it does not enter the nucleus, which avoids its integration into the genome and the risk of insertional mutagenesis. The large-scale production of IVT mRNA is less complex than production of recombinant proteins, and Good Manufacturing Practice-compliant mRNA production is easily scalable, ideally poising mRNA for not only off-the-shelf, but more personalized treatment approaches. IVT mRNA's safety profile, pharmacokinetics, and pharmacodynamics, including its inherent immunostimulatory capacity, can be optimized for different therapeutic applications by harnessing a wide array of optimized sequence elements, chemical modifications, purification techniques, and delivery methods. The value of IVT mRNA was recently proved during the COVID-19 pandemic when mRNA-based vaccines outperformed the efficacy of established technologies, and millions of doses were rapidly deployed. In this review, we will discuss chemical modifications of IVT mRNA and highlight numerous preclinical and clinical applications including vaccines for cancer and infectious diseases, cancer immunotherapy, protein replacement, gene editing, and cell reprogramming.
Collapse
|
76
|
Di Trani CA, Cirella A, Arrizabalaga L, Fernandez-Sendin M, Bella A, Aranda F, Melero I, Berraondo P. Overcoming the limitations of cytokines to improve cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:107-141. [PMID: 35777862 DOI: 10.1016/bs.ircmb.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytokines are pleiotropic soluble proteins used by immune cells to orchestrate a coordinated response against pathogens and malignancies. In cancer immunotherapy, cytokine-based drugs can be developed potentiating pro-inflammatory cytokines or blocking immunosuppressive cytokines. However, the complexity of the mechanisms of action of cytokines requires the use of biotechnological strategies to minimize systemic toxicity, while potentiating the antitumor response. Sequence mutagenesis, fusion proteins and gene therapy strategies are employed to enhance the half-life in circulation, target the desired bioactivity to the tumor microenvironment, and to optimize the therapeutic window of cytokines. In this review, we provide an overview of the different strategies currently being pursued in pre-clinical and clinical studies to make the most of cytokines for cancer immunotherapy.
Collapse
Affiliation(s)
- Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Leire Arrizabalaga
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Angela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
77
|
Atallah-Yunes SA, Robertson MJ. Cytokine Based Immunotherapy for Cancer and Lymphoma: Biology, Challenges and Future Perspectives. Front Immunol 2022; 13:872010. [PMID: 35529882 PMCID: PMC9067561 DOI: 10.3389/fimmu.2022.872010] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/22/2022] [Indexed: 11/15/2022] Open
Abstract
Cytokines regulate both the innate and adaptive immune responses to cancer. Although antitumor activity has been seen for several cytokines in preclinical models, they have had limited success as single therapeutic agents in clinical trials of cancer immunotherapy. However, the possible combinations of cytokines with other immune therapeutics and the advancement in genetic engineering, synthetic biology and cellular and immune therapy has led to the revival of interest in cytokines as anticancer agents. This article will review several immunostimulatory cytokines with anticancer activity, focusing on the those that have been studied in treatment of lymphoma and highlighting recent advances of potential clinical relevance.
Collapse
Affiliation(s)
- Suheil Albert Atallah-Yunes
- Department of Hematology and Medical Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michael J Robertson
- Department of Hematology and Medical Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
78
|
van Akkooi ACJ, Zijlker LP, Wouters MWJM. Neoadjuvant Immune Checkpoint Inhibitor Therapy in Melanoma: Efficacy, Safety and Timing. BioDrugs 2022; 36:373-380. [PMID: 35397089 PMCID: PMC9148869 DOI: 10.1007/s40259-022-00525-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 11/27/2022]
Abstract
The introduction of effective systemic therapies has significantly changed the treatment of stage III and IV melanoma. Both immune checkpoint inhibitors and targeted therapies have improved recurrence-free survival in the adjuvant setting. Recent interest has sparked for neoadjuvant systemic therapy with immune checkpoint inhibitors. The intended benefit of pre-operative treatment with immunotherapy is amongst others to enable tailoring of the surgery and adjuvant systemic therapy according to the treatment response. Most importantly, recurrence-free survival might be improved by neoadjuvant systemic therapy over the current standard of care of surgery followed by adjuvant systemic therapy. The first phase I and II trials investigating anti-PD1 inhibitors, both as a single agent and in combination with anti-CTLA-4 inhibitors or other therapeutic agents, have shown promising results. Pathological complete response on neoadjuvant systemic therapy seems a valid surrogate endpoint for relapse-free and overall survival. Pathological complete response rates in these trials vary between 30 and 70%. The optimal dose with respect to efficacy and toxicity and the interval between systemic and surgical treatment remain important issues to address. Accumulating follow-up data and ongoing phase III studies must prove if neoadjuvant systemic therapy is superior to surgery followed by standard-of-care adjuvant therapy.
Collapse
Affiliation(s)
- Alexander C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Crows Nest, PO Box 1479, Sydney, NSW, 1585, Australia.
- Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | | | - Michel W J M Wouters
- Netherlands Cancer Institute, Amsterdam, The Netherlands
- Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
79
|
Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol 2022; 19:237-253. [PMID: 34997230 DOI: 10.1038/s41571-021-00588-9] [Citation(s) in RCA: 526] [Impact Index Per Article: 175.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 12/14/2022]
Abstract
During the past 40 years, cytokines and cytokine receptors have been extensively investigated as either cancer targets or cancer treatments. A strong preclinical rationale supports therapeutic strategies to enhance the growth inhibitory and immunostimulatory effects of interferons and interleukins, including IL-2, IL-7, IL-12 and IL-15, or to inhibit the inflammatory and tumour-promoting actions of cytokines such as TNF, IL-1β and IL-6. This rationale is underscored by the discovery of altered and dysregulated cytokine expression in all human cancers. These findings prompted clinical trials of several cytokines or cytokine antagonists, revealing relevant biological activity but limited therapeutic efficacy. However, most trials involved patients with advanced-stage disease, which might not be the optimal setting for cytokine-based therapy. The advent of more effective immunotherapies and an increased understanding of the tumour microenvironment have presented new approaches to harnessing cytokine networks in the treatment of cancer, which include using cytokine-based therapies to enhance the activity or alleviate the immune-related toxicities of other treatments as well as to target early stage cancers. Many challenges remain, especially concerning delivery methods, context dependencies, and the pleiotropic, redundant and often conflicting actions of many cytokines. Herein, we discuss the lessons learnt from the initial trials of single-agent cytokine-based therapies and subsequent efforts to better exploit such agents for the treatment of solid tumours.
Collapse
Affiliation(s)
- David J Propper
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Frances R Balkwill
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
80
|
Ernst M, Giubellino A. The Current State of Treatment and Future Directions in Cutaneous Malignant Melanoma. Biomedicines 2022; 10:822. [PMID: 35453572 PMCID: PMC9029866 DOI: 10.3390/biomedicines10040822] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Malignant melanoma is the leading cause of death among cutaneous malignancies. While its incidence is increasing, the most recent cancer statistics show a small but clear decrease in mortality rate. This trend reflects the introduction of novel and more effective therapeutic regimens, including the two cornerstones of melanoma therapy: immunotherapies and targeted therapies. Immunotherapies exploit the highly immunogenic nature of melanoma by modulating and priming the patient's own immune system to attack the tumor. Treatments combining immunotherapies with targeted therapies, which disable the carcinogenic products of mutated cancer cells, have further increased treatment efficacy and durability. Toxicity and resistance, however, remain critical challenges to the field. The present review summarizes past treatments and novel therapeutic interventions and discusses current clinical trials and future directions.
Collapse
Affiliation(s)
| | - Alessio Giubellino
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
81
|
Szatkowska L, Sieczek J, Tekiela K, Ziętek M, Stachyra-Strawa P, Cisek P, Matkowski R. Outcomes of Patients with Metastatic Melanoma-A Single-Institution Retrospective Analysis. Cancers (Basel) 2022; 14:cancers14071672. [PMID: 35406444 PMCID: PMC8997072 DOI: 10.3390/cancers14071672] [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: 02/24/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Background: This study assessed risk factors and the results of treatment with anti-PD-1 antibodies and BRAF/MEK inhibitors for advanced malignant melanoma. Methods: A retrospective analysis was performed on 52 patients treated with immunotherapy and BRAF/MEK inhibitors for disseminated malignant melanoma. Results: The median follow-up was 31 months (6−108 months). The median PFS1 was 6 months (1−44 months). Second-line systemic treatment was applied in 27 patients (52%). The median PFS2 was 2 months (0−27 months), and the median OS was 31 months (6−108 months). Among the analyzed risk factors, only the presence of the BRAF mutation was statistically significant for disease recurrence after surgery. In patients undergoing anti-BRAF/MEK therapy, the median PFS1 was 7 months, and in patients undergoing mono-immunotherapy, 4 months. The 12- and 24-month PFS1 rates in the group treated with BRAF inhibitors were 29 and 7%, respectively, and in patients treated with mono-immunotherapy 13 and 0%, respectively (Z = 1.998, p = 0.04). The type of treatment used had no effect on OS (Z = 0.237, p > 0.05). Conclusion: Patients with the V600 mutation should be closely monitored. In the event of disease recurrence, treatment with BRAF/MEK inhibitors should be considered. The type of treatment used has no effect on OS.
Collapse
Affiliation(s)
- Lidia Szatkowska
- Clinical Department of Cardiology, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland
- Correspondence:
| | - Jan Sieczek
- Department of Orthopedic Surgery, Provincial Specialist Hospital, Kamieńskiego 73A, 51-124 Wrocław, Poland;
| | - Katarzyna Tekiela
- Department of Oncology, Lower Silesian Oncology, Pulmonology and Hematology Center, Plac Hirszfelda 12, 53-413 Wrocław, Poland;
| | - Marcin Ziętek
- Department of Oncology, Wrocław Medical University, wyb. L. Pasteura 1, 50-367 Wrocław, Poland; (M.Z.); (R.M.)
- Department of Surgical Oncology, Lower Silesian Oncology, Pulmonology and Hematology Center, Plac Hirszfelda 12, 53-413 Wrocław, Poland
| | - Paulina Stachyra-Strawa
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland; (P.S.-S.); (P.C.)
| | - Paweł Cisek
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland; (P.S.-S.); (P.C.)
| | - Rafał Matkowski
- Department of Oncology, Wrocław Medical University, wyb. L. Pasteura 1, 50-367 Wrocław, Poland; (M.Z.); (R.M.)
- Department of Surgical Oncology, Lower Silesian Oncology, Pulmonology and Hematology Center, Plac Hirszfelda 12, 53-413 Wrocław, Poland
| |
Collapse
|
82
|
Recent Advances and Next Breakthrough in Immunotherapy for Cancer Treatment. J Immunol Res 2022; 2022:8052212. [PMID: 35340585 PMCID: PMC8956433 DOI: 10.1155/2022/8052212] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
With the huge therapeutic potential, cancer immunotherapy is expected to become the mainstream of cancer treatment. In the current field of cancer immunotherapy, there are mainly five types. Immune checkpoint blockade therapy is one of the most promising directions. Adoptive cell therapy is an important component of cancer immunotherapy. The therapy with the cancer vaccine is promising cancer immunotherapy capable of cancer prevention. Cytokine therapy is one of the pillars of cancer immunotherapy. Oncolytic immunotherapy is a promising novel component of cancer immunotherapy, which with significantly lower incidence of serious adverse reactions. The recent positive results of many clinical trials with cancer immunotherapy may herald good clinical prospects. But there are still many challenges in the broad implementation of immunotherapy. Such as the immunotherapy cannot act on all tumors, and it has serious adverse effects including but not limited to nonspecific and autoimmunity inflammation. Here, we center on recent progress made within the last 5 years in cancer immunotherapy. And we discuss the theoretical background, as well as the opportunities and challenges of cancer immunotherapy.
Collapse
|
83
|
Witt RG, Erstad DJ, Wargo JA. Neoadjuvant therapy for melanoma: rationale for neoadjuvant therapy and pivotal clinical trials. Ther Adv Med Oncol 2022; 14:17588359221083052. [PMID: 35251322 PMCID: PMC8894940 DOI: 10.1177/17588359221083052] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/03/2022] [Indexed: 12/26/2022] Open
Abstract
The treatment of malignant melanoma has drastically changed over the past decade with the advent of immune checkpoint blockade, targeted therapy with BRAF/MEK inhibition, and other novel therapies such as oncolytic virus intralesional therapy. Despite improvements in patient response rates and survival with these new treatments, there exists a large portion of patients with surgically resectable disease that are high risk for relapse. Patients with high-risk resectable melanoma account for up to 20% of newly diagnosed cases. For this high-risk group of patients, neoadjuvant therapy has many purposed advantages over adjuvant therapy, including a more robust immune response due to abundant tumor antigens at treatment initiation, the ability to assess pathologic response to therapy, tumor downstaging leading to increased disease resectability, and a potential decreased need for extensive lymphadenectomies. These findings have been backed by preclinical models and multiple neoadjuvant trials are underway. In this review, we will discuss the trials that have set the foundation for the current treatment standards and discuss the role and rationale for neoadjuvant therapy for high-risk malignant melanomas.
Collapse
Affiliation(s)
- Russell G. Witt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Derek J. Erstad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1484, Houston, TX 77030-4009, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
84
|
Grossmann KF, Othus M, Patel SP, Tarhini AA, Sondak VK, Knopp MV, Petrella TM, Truong TG, Khushalani NI, Cohen JV, Buchbinder EI, Kendra K, Funchain P, Lewis KD, Conry RM, Chmielowski B, Kudchadkar RR, Johnson DB, Li H, Moon J, Eroglu Z, Gastman B, Kovacsovics-Bankowski M, Gunturu KS, Ebbinghaus SW, Ahsan S, Ibrahim N, Sharon E, Korde LA, Kirkwood JM, Ribas A. Adjuvant Pembrolizumab versus IFNα2b or Ipilimumab in Resected High-Risk Melanoma. Cancer Discov 2022; 12:644-653. [PMID: 34764195 PMCID: PMC8904282 DOI: 10.1158/2159-8290.cd-21-1141] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/19/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
We conducted a randomized phase III trial to evaluate whether adjuvant pembrolizumab for one year (647 patients) improved recurrence-free survival (RFS) or overall survival (OS) in comparison with high-dose IFNα-2b for one year or ipilimumab for up to three years (654 patients), the approved standard-of-care adjuvant immunotherapies at the time of enrollment for patients with high-risk resected melanoma. At a median follow-up of 47.5 months, pembrolizumab was associated with significantly longer RFS than prior standard-of-care adjuvant immunotherapies [HR, 0.77; 99.62% confidence interval (CI), 0.59-0.99; P = 0.002]. There was no statistically significant association with OS among all patients (HR, 0.82; 96.3% CI, 0.61-1.09; P = 0.15). Proportions of treatment-related adverse events of grades 3 to 5 were 19.5% with pembrolizumab, 71.2% with IFNα-2b, and 49.2% with ipilimumab. Therefore, adjuvant pembrolizumab significantly improved RFS but not OS compared with the prior standard-of-care immunotherapies for patients with high-risk resected melanoma. SIGNIFICANCE Adjuvant PD-1 blockade therapy decreases the rates of recurrence, but not survival, in patients with surgically resectable melanoma, substituting the prior standard-of-care immunotherapies for this cancer. See related commentary by Smithy and Shoushtari, p. 599. This article is highlighted in the In This Issue feature, p. 587.
Collapse
Affiliation(s)
| | - Megan Othus
- SWOG Statistics and Data Management Center, Seattle
| | - Sapna P. Patel
- The University of Texas MD Anderson Cancer Center, Houston
| | | | | | | | | | | | | | - Justine V. Cohen
- Massachusetts General Hospital, Boston (during conduct of trial), University of Pennsylvania, Philadelphia (current)
| | | | | | | | | | - Robert M. Conry
- University of Alabama at Birmingham Cancer Center, Birmingham (during conduct of trial), Clearview Cancer Institute, Anniston (current)
| | - Bartosz Chmielowski
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles
| | | | | | - Hongli Li
- SWOG Statistics and Data Management Center, Seattle
| | - James Moon
- SWOG Statistics and Data Management Center, Seattle
| | - Zeynep Eroglu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | | | | | | | | | | | | | - Elad Sharon
- National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda
| | - Larissa A. Korde
- National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda
| | | | - Antoni Ribas
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles
| |
Collapse
|
85
|
Smithy JW, Shoushtari AN. Adjuvant PD-1 Blockade in Resected Melanoma: Is Preventing Recurrence Enough? Cancer Discov 2022; 12:599-601. [PMID: 35257151 DOI: 10.1158/2159-8290.cd-21-1593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
SUMMARY Grossmann and colleagues report the results of a large randomized trial demonstrating improved recurrence-free survival with adjuvant pembrolizumab in resected melanoma compared with adjuvant ipilimumab or IFNα2b. However, it remains unclear whether adjuvant immunotherapies extend overall survival as outcomes for patients with advanced melanoma continue to improve. See related article by Grossmann et al., p. 644 (1).
Collapse
Affiliation(s)
- James W Smithy
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
86
|
Truong TG, Kennedy LB, Patel SP. 25 Years of Adjuvant Therapy in Melanoma: A Perspective on Current Approvals and Insights into Future Directions. Curr Oncol Rep 2022; 24:533-542. [PMID: 35192117 DOI: 10.1007/s11912-022-01232-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Thach-Giao Truong
- Kaiser Permanente Northern California, 975 Sereno Drive, Vallejo, CA, 94589, USA
| | - Lucy Boyce Kennedy
- The Cleveland Clinic Foundation, 9500 Euclid Ave CA-60, Cleveland, OH, 44195, USA
| | - Sapna P Patel
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0430, Houston, TX, 77030, USA.
| |
Collapse
|
87
|
Drescher NR, Amdur RJ, Morris CG, Shaw CM, Dziegielewski PT, Mendenhall WM. Postoperative Radiotherapy for Cutaneous Melanoma in Patients at High Risk of Local-Regional Recurrence after Surgery Alone. Cancer Invest 2022; 40:348-353. [PMID: 35100059 DOI: 10.1080/07357907.2022.2033986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
We evaluated outcomes in 131 patients with cutaneous melanoma (median follow-up, 3.6 years) considered at high risk of recurrence after surgery alone treated with surgery and postoperative radiotherapy. Eligible patients had one or more of the following: recurrence after surgery, positive lymph nodes, extracapsular extension, incomplete regional node dissection, microscopically positive margins, gross residual disease, or in-transit metastases. 102 patients received hypofractionated radiotherapy and 29 had conventional fractionation. 10-year outcomes were: in-field local-regional control, 87%; local regional control, 72%; distant metastasis-free survival, 48%; cause-specific survival, 44%; and overall survival, 31%. Three patients experienced acute toxicities while 6 experienced late toxicities.
Collapse
Affiliation(s)
- Nicolette R Drescher
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Robert J Amdur
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christopher G Morris
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christiana M Shaw
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Peter T Dziegielewski
- Department of Otolaryngology, University of Florida College of Medicine, Gainesville, FL, USA
| | - William M Mendenhall
- Department of Radiation Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
88
|
Scott AM, Lee ST, Senko C, Ciprotti M, Kee D. Diagnostic Applications of Nuclear Medicine: Malignant Melanoma. NUCLEAR ONCOLOGY 2022:1235-1269. [DOI: 10.1007/978-3-031-05494-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
89
|
Ahmad Z, Somanath PR. AKT Isoforms in the Immune Response in Cancer. Curr Top Microbiol Immunol 2022; 436:349-366. [PMID: 36243852 DOI: 10.1007/978-3-031-06566-8_15] [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: 06/16/2023]
Abstract
AKT is a protein kinase that exists in three isoforms: AKT1, AKT2, and AKT3. Though similar in structure, these isoforms display different effects. AKT is activated downstream of PI3K, and together, this signaling pathway helps regulate cellular processes including cell growth, proliferation, metabolism, survival, and apoptosis. Disruption in these pathways has been associated with disorders including cardiovascular diseases, developmental disorders, inflammatory responses, autoimmune diseases, neurologic disorders, type 2 diabetes, and several cancers. In cancer, deregulation in the PI3K/AKT pathway can be manifested as tumorigenesis, pathological angiogenesis, and metastasis. Increased activity has been correlated with tumor progression and resistance to cancer treatments. Recent studies have suggested that inhibition of the PI3K/AKT pathway plays a significant role in the development, expansion, and proliferation of cells of the immune system. Additionally, AKT has been found to play an important role in differentiating regulatory T cells, activating B cells, and augmenting tumor immunosurveillance. This emphasizes AKT as a potential target for inhibition in cancer therapy. This chapter reviews AKT structure and regulation, its different isoforms, its role in immune cells, and its modulation in oncotherapy.
Collapse
Affiliation(s)
- Zayd Ahmad
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Georgia Cancer Center, Vascular Biology Center and Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
90
|
Tuli HS, Sak K, Iqubal A, Choudhary R, Adhikary S, Kaur G, Kumar P, Garg VK. Recent Advances in Immunotherapy for the Treatment of Malignant Melanoma. Curr Pharm Des 2022; 28:2363-2374. [PMID: 35894458 DOI: 10.2174/1381612828666220727124639] [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: 11/09/2021] [Accepted: 05/04/2022] [Indexed: 11/22/2022]
Abstract
Malignant melanoma is one of the most worrisome malignancies due to its fast dissemination and early formation of metastases in multiple sites throughout the body. Despite the intensive efforts made in the last decades, the use of standard therapeutic strategies, including chemotherapy and radiotherapy, has not led to a substantial improvement in clinical outcomes, mainly because of the intrinsic resistance of melanomas to these treatment modalities. Therefore, in recent years, numerous studies have focused on the possibility of boosting the clinical responses of melanoma patients by using novel immunotherapeutic agents. In this review article, a comprehensive survey is presented about the approved immunotherapeutic drugs and their action mechanisms, besides describing the agents that are currently still in clinical trials. Moreover, the combination of immunotherapeutic drugs with conventional approaches, i.e., radiotherapy, chemotherapy, and targeted therapy, is another focal point of this review, providing valuable input for further elaboration of the best treatment regimens to prolong survival and improve the quality of life of melanoma patients.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | | | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (Formerly Faculty of Pharmacy), Jamia Hamdard (Deemed to be University), Delhi, India
| | - Renuka Choudhary
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Shubham Adhikary
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | - Pawan Kumar
- Institute of Plant Sciences, Agricultural Research Organisation (ARO), 7505101, Rishon LeZion, Israel
| | - Vivek Kumar Garg
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University, Gharuan, Mohali - 140413, Punjab, India
| |
Collapse
|
91
|
Apostu RC, Stefanescu E, Scurtu RR, Kacso G, Drasovean R. Difficulties in diagnosing anorectal melanoma: A case report and review of the literature. World J Clin Cases 2021; 9:11369-11381. [PMID: 35071568 PMCID: PMC8717525 DOI: 10.12998/wjcc.v9.i36.11369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anorectal melanoma is a tumour that is difficult to identify due to its rarity and variability of presentation. Insufficient data published in the literature do not allow for diagnostic and treatment guidelines to be established. Anorectal melanoma has the worst prognosis among mucosal melanomas and is frequently misdiagnosed by standard identification methods.
CASE SUMMARY A 66-year-old woman presented with intermittent anal bleeding, pain, and tenesmus in the past month, with no associated weight loss. Colonoscopy revealed a cauliflower-like tumour with a diameter of 1.5 cm, with exulcerated areas and an adherent clot but without obstruction. Biopsy results identified an inflammatory rectal polyp with nonspecific chronic rectitis. Tumour markers CA 19-9 and CEA were within the normal range. After 6 mo, due to the persistence of symptoms, a pelvic magnetic resonance imaging scan was performed. A lesion measuring 2.8 cm × 2.7 cm × 2.1 cm was identified at the anorectal junction, along with two adjacent lymphadenopathies. No distant metastases were detected. Immunohistochemistry was performed on the second set of biopsies, and a diagnosis of anorectal melanoma was established. Surgical treatment by abdominoperineal resection was performed. Evolution was marked by the appearance of lung metastases at 1 mo postoperatively, detected on a positron emission tomography-computer tomography scan, and perineal recurrence after 5 mo. After molecular testing, the patient was included in an immunotherapy trial.
CONCLUSION This case highlights the difficulty of establishing a definitive early diagnosis of anorectal melanoma, the importance of performing histological analysis on a well-represented biopsy specimen, and the poor prognosis, even with radical surgery.
Collapse
Affiliation(s)
- Raluca Cristina Apostu
- Department of Surgery, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca 400006, Romania
| | - Elena Stefanescu
- Department of Anesthesia and Intensive Care, Emergency County Clinical Hospital, Cluj-Napoca 400006, Romania
| | - Radu Razvan Scurtu
- Department of Surgery, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Cluj Napoca 400006, Romania
| | - Gabriel Kacso
- Department of Medical Oncology and Radiotherapy, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca 400006, Romania
| | - Radu Drasovean
- Department of Surgery, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca 400006, Romania
| |
Collapse
|
92
|
Lian G, Mak TSK, Yu X, Lan HY. Challenges and Recent Advances in NK Cell-Targeted Immunotherapies in Solid Tumors. Int J Mol Sci 2021; 23:164. [PMID: 35008589 PMCID: PMC8745474 DOI: 10.3390/ijms23010164] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cell is a powerful malignant cells killer, providing rapid immune responses via direct cytotoxicity without the need of antigen processing and presentation. It plays an essential role in preventing early tumor, metastasis and minimal residual disease. Although adoptive NK therapies achieved great success in clinical trials against hematologic malignancies, their accumulation, activation, cytotoxic and immunoregulatory functions are severely impaired in the immunosuppressive microenvironment of solid tumors. Now with better understandings of the tumor evasive mechanisms from NK-mediated immunosurveillance, immunotherapies targeting the key molecules for NK cell dysfunction and exhaustion have been developed and tested in both preclinical and clinical studies. In this review, we introduce the challenges that NK cells encountered in solid tumor microenvironment (TME) and the therapeutic approaches to overcome these limitations, followed by an outline of the recent preclinical advances and the latest clinical outcomes of NK-based immunotherapies, as well as promising strategies to optimize current NK-targeted immunotherapies for solid tumors.
Collapse
Affiliation(s)
- Guangyu Lian
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China;
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Thomas Shiu-Kwong Mak
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Xueqing Yu
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China;
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Hui-Yao Lan
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
93
|
Dizman N, Buchbinder EI. Cancer Therapy Targeting CD47/SIRPα. Cancers (Basel) 2021; 13:cancers13246229. [PMID: 34944850 PMCID: PMC8699673 DOI: 10.3390/cancers13246229] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 01/01/2023] Open
Abstract
Simple Summary The interaction between cluster of differentiation 47 (CD47) on cancer cells and signal regulatory protein alpha (SIRPα) on immune cells, such as macrophages and dendritic cells, generates a “don’t eat me” signal. This is a common mechanism that provides cancer cells an escape from the innate immune system. Several therapeutics directed to CD47 or SIRPα have entered early clinical trials in recent years. In this article, we review the role of CD47/SIRPα axis in cancer, and summarize the literature on the efficacy and safety of therapeutics targeting CD47 or SIRPα. We also discuss the future implementation of these therapeutics in the treatments of various cancer types. Abstract In the past decade, the field of cancer immunotherapy has rapidly advanced, establishing a crucial role for immune checkpoint blockers in the treatment of a variety of cancer types. In parallel with these remarkable clinical developments, further efforts have focused on ways of unleashing adaptive immune responses against cancer. CD47, a cell surface molecule overexpressed by several cancer types that facilitates immune escape from macrophages, dendritic cells and natural killer cells, and its ligand SIRPα, have emerged as potential therapeutic targets. A number of agents directed to CD47/SIRPα have been developed and demonstrated preclinical activity. Early phase clinical trials are investigating CD47/SIRPα directed agents with available data, suggesting safety and preliminary activity. Herein, we provide an overview of the mechanistic rationale of targeting CD47/SIRPα axis and associated clinical evidence.
Collapse
Affiliation(s)
- Nazli Dizman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Elizabeth I. Buchbinder
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- Correspondence:
| |
Collapse
|
94
|
Mao C, Ding Y, Xu N. A Double-Edged Sword Role of Cytokines in Prostate Cancer Immunotherapy. Front Oncol 2021; 11:688489. [PMID: 34868907 PMCID: PMC8635015 DOI: 10.3389/fonc.2021.688489] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer (PC) is one of the most common malignancies among men and is the second leading cause of cancer death. PC immunotherapy has taken relatively successful steps in recent years, and these treatments are still being developed and tested. Evidence suggests that immunotherapy using cytokines as essential mediators in the immune system may help treat cancer. It has been shown that cytokines play an important role in anti-tumor defense. On the other hand, other cytokines can also favor the tumor and suppress anti-tumor responses. Moreover, the dose of cytokine in cancer cytokine-based immunotherapy, as well as the side effects of high doses, can also affect the outcomes of treatment. Cytokines can also be determinative in the outcome of other immunotherapy methods used in PC. In this review, the role of cytokines in the pathogenesis of cancer and their impacts on the main types of immunotherapies in the treatment of PC are discussed.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Medical Oncology Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nong Xu
- Department of Medical Oncology Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
95
|
Davis L, Tarduno A, Lu YC. Neoantigen-Reactive T Cells: The Driving Force behind Successful Melanoma Immunotherapy. Cancers (Basel) 2021; 13:cancers13236061. [PMID: 34885172 PMCID: PMC8657037 DOI: 10.3390/cancers13236061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Cancer immunotherapy is a revolutionary type of cancer therapy. It uses the patient’s own immune system to fight and potentially cure cancer. The first major breakthrough of immunotherapy came from successful clinical trials for melanoma treatments. Since then, researchers have focused on understanding the science behind immunotherapy, so that patients with other types of cancer may also benefit. One of the major findings is that the T cells in melanoma patients may recognize a specific type of tumor antigen, called neoantigens, and then kill tumor cells that present these neoantigens. The neoantigens mainly arise from the DNA mutations found in tumor cells. These mutations are translated into mutated proteins that are then distinguished by T cells. In this article, we discuss the critical role of T cells in immunotherapy, as well as the clinical trials that shaped the treatments for melanoma. Abstract Patients with metastatic cutaneous melanoma have experienced significant clinical responses after checkpoint blockade immunotherapy or adoptive cell therapy. Neoantigens are mutated proteins that arise from tumor-specific mutations. It is hypothesized that the neoantigen recognition by T cells is the critical step for T-cell-mediated anti-tumor responses and subsequent tumor regressions. In addition to describing neoantigens, we review the sentinel and ongoing clinical trials that are helping to shape the current treatments for patients with cutaneous melanoma. We also present the existing evidence that establishes the correlations between neoantigen-reactive T cells and clinical responses in melanoma immunotherapy.
Collapse
Affiliation(s)
- Lindy Davis
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Ashley Tarduno
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Yong-Chen Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| |
Collapse
|
96
|
Amabile S, Roccuzzo G, Pala V, Tonella L, Rubatto M, Merli M, Fava P, Ribero S, Fierro MT, Queirolo P, Quaglino P. Clinical Significance of Distant Metastasis-Free Survival (DMFS) in Melanoma: A Narrative Review from Adjuvant Clinical Trials. J Clin Med 2021; 10:jcm10235475. [PMID: 34884176 PMCID: PMC8658595 DOI: 10.3390/jcm10235475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Cutaneous melanoma is the most dangerous skin cancer, with high death rates in advanced stages. To assess the impact of each treatment on patient outcomes, most studies use relapse-free survival (RFS) as a primary endpoint and distant metastasis-free survival (DMFS) as a secondary endpoint. The aim of this narrative review of the main adjuvant studies for resected stage III/IV melanoma, with a specific focus on DMFS, is to evaluate DMFS trends and their potential association with RFS, identify which treatments are possibly associated with better outcomes in terms of DMFS and their potential predictive factors, and discuss DMFS trends in terms of patient management in daily practice. We outline the impact of each available treatment option on DMFS and RFS according to the years of follow-up and compare data from different studies. Overall, the trends of DMFS closely follow those of RFS, with most patients relapsing at visceral rather than regional sites. As it captures the burden of patients who develop distant relapse, DMFS could be considered a primary endpoint, in addition to RFS, in adjuvant trials, identifying patients whose relapse is associated with a worse prognosis and who may need further systemic treatment.
Collapse
Affiliation(s)
- Simone Amabile
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Gabriele Roccuzzo
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
- Correspondence: ; Tel.: +39-01-1633-5843
| | - Valentina Pala
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Luca Tonella
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Marco Rubatto
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Martina Merli
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Paolo Fava
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Simone Ribero
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Maria Teresa Fierro
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| | - Paola Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, European Institute of Oncology (IEO), European Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20141 Milan, Italy;
| | - Pietro Quaglino
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (S.A.); (V.P.); (L.T.); (M.R.); (M.M.); (P.F.); (S.R.); (M.T.F.); (P.Q.)
| |
Collapse
|
97
|
Ott PA. Capitalizing on the messenger: Intra-tumoral delivery of RNA with a systemic effect. Cancer Cell 2021; 39:1458-1460. [PMID: 34752754 PMCID: PMC9261938 DOI: 10.1016/j.ccell.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
As reported recently in Science Translational Medicine, intra-tumoral administration of mRNAs that encode four cytokines formulated in saline mediates regression of injected and distant murine tumors. The anti-tumor effect, which is also seen in the anti-PD-1-resistant setting and enhanced by checkpoint blockade, is mediated by tumor-specific T cells and natural killer cells.
Collapse
Affiliation(s)
- Patrick A Ott
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
98
|
Nguyen MHT, Luo YH, Li AL, Tsai JC, Wu KL, Chung PJ, Ma N. miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma. Biomolecules 2021; 11:1648. [PMID: 34827646 PMCID: PMC8615556 DOI: 10.3390/biom11111648] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint inhibitors are a promising therapy for the treatment of cancers, including melanoma, that improved benefit clinical outcomes. However, a subset of melanoma patients do not respond or acquire resistance to immunotherapy, which limits their clinical applicability. Recent studies have explored the reasons related to the resistance of melanoma to immune checkpoint inhibitors. Of note, miRNAs are the regulators of not only cancer progression but also of the response between cancer cells and immune cells. Investigation of miRNA functions within the tumor microenvironment have suggested that miRNAs could be considered as key partners in immunotherapy. Here, we reviewed the known mechanism by which melanoma induces resistance to immunotherapy and the role of miRNAs in immune responses and the microenvironment.
Collapse
Affiliation(s)
- Mai-Huong Thi Nguyen
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.N.); (A.-L.L.); (K.-L.W.); (P.-J.C.)
| | - Yueh-Hsia Luo
- Department of Life Sciences, National Central University, Taoyuan 320317, Taiwan;
| | - An-Lun Li
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.N.); (A.-L.L.); (K.-L.W.); (P.-J.C.)
| | - Jen-Chieh Tsai
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 300044, Taiwan;
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Kun-Lin Wu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.N.); (A.-L.L.); (K.-L.W.); (P.-J.C.)
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 325208, Taiwan
| | - Pei-Jung Chung
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.N.); (A.-L.L.); (K.-L.W.); (P.-J.C.)
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.N.); (A.-L.L.); (K.-L.W.); (P.-J.C.)
| |
Collapse
|
99
|
Molecular Analysis of Elements of Melanoma Insensitivity to TCR-Engineered Adoptive Cell Therapy. Int J Mol Sci 2021; 22:ijms222111726. [PMID: 34769156 PMCID: PMC8584081 DOI: 10.3390/ijms222111726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
Metastatic melanoma accounts for the highest number of skin cancer-related deaths. Traditional treatments are ineffective due to their inability to induce tumor regression at a high rate. Newer treatments such as immune checkpoint inhibitors (ICI), targeted therapy (BRAFi and MEKi), and T cell receptor (TCR)-engineered T cells aim to increase the ability of the host immune system to recognize and eradicate tumors. ICIs inhibit negative regulatory mechanisms and boost the antitumor activity of the host’s immune system, while targeted therapy directed against aberrant signaling molecules (BRAF and MEK) will block the uncontrolled proliferation and expansion of melanomas. The basis of the TCR-engineered T cell strategy is to transduce host T cells with antigen-specific TCRα/β chains to produce high-affinity T cells for tumor-associated antigens. TCR-transgenic T cells are expanded and activated ex vivo and reinfused into patients to increase the targeting of cancer cells. While these treatments have had varyingly favorable results, their efficacy is limited due to inherent or acquired resistance. Various mechanisms explain melanoma immune-resistance, including the loss or downregulation of the MCH/peptide complex, aberrant activity of signaling pathways, and altered dynamics of apoptotic machinery. Collectively, these mechanisms confer melanoma resistance to apoptotic stimuli delivered by T cells despite a fully functional and effective antitumor immune response. Identification of biomarkers, combination treatment, and the use of CAR T cells are among the approaches that can potentially circumvent melanoma’s resistance to immunotherapy.
Collapse
|
100
|
de Meira Júnior JD, Sobrado LF, Guzela VM, Nahas SC, Sobrado CW. Anorectal Mucosal Melanoma: A Case Report and Literature Review. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e933032. [PMID: 34699518 PMCID: PMC8557858 DOI: 10.12659/ajcr.933032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Anorectal mucosal melanoma (AMM) is a rare and aggressive neoplasm, with a 5-year survival rate of 10%. Due to its rarity and nonspecific symptoms, the diagnosis is often made late. Surgical resection remains the criterion standard for treatment of anorectal melanoma. CASE REPORT We present the case of an 81-year-old woman presenting with hematochezia, anal secretion, tenesmus, difficulty in defecation, and perianal pain. On physical examination, there was a prolapse of a 5-cm melanocytic nodule in the anal canal, hard on palpation. Biopsy confirmed anorectal melanoma. Staging revealed anal and metastatic disease, with adrenal, lymphatic, and hepatic involvement. As the patient continued to have bleeding, severe pain, and difficulty in defecation, she was submitted to a wide local excision. At 5-month follow-up, the anal lesion had relapsed, and the patient died 10 months after the procedure. CONCLUSIONS AMM is a rare and extremely aggressive tumor. Symptoms are nonspecific but early diagnosis should be pursued to allow curative treatment. Surgical resection with free margins is the goal of surgical treatment. New therapies are being studied, including immunotherapy, which can improve the dismal prognosis of this rare disease.
Collapse
Affiliation(s)
- José Donizeti de Meira Júnior
- Colorectal Surgery Division, Department of Gastroenterology, Clinical Hospital, University of São Paulo - School of Medicine, São Paulo, SP, Brazil
| | - Lucas Faraco Sobrado
- Colorectal Surgery Division, Department of Gastroenterology, Clinical Hospital, University of São Paulo - School of Medicine, São Paulo, SP, Brazil
| | - Vivian M Guzela
- Colorectal Surgery Division, Department of Gastroenterology, Clinical Hospital, University of São Paulo - School of Medicine, São Paulo, SP, Brazil
| | - Sergio Carlos Nahas
- Colorectal Surgery Division, Department of Gastroenterology, Clinical Hospital, University of São Paulo - School of Medicine, São Paulo, SP, Brazil
| | - Carlos Walter Sobrado
- Colorectal Surgery Division, Department of Gastroenterology, Clinical Hospital, University of São Paulo - School of Medicine, São Paulo, SP, Brazil
| |
Collapse
|