1
|
Gujar H, Mehta A, Li HT, Tsai YC, Qiu X, Weisenberger DJ, Jasiulionis MG, In GK, Liang G. Characterizing DNA methylation signatures and their potential functional roles in Merkel cell carcinoma. Genome Med 2021; 13:130. [PMID: 34399838 PMCID: PMC8365948 DOI: 10.1186/s13073-021-00946-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Merkel cell carcinoma (MCC) is a rare but aggressive skin cancer with limited treatment possibilities. Merkel cell tumors display with neuroendocrine features and Merkel cell polyomavirus (MCPyV) infection in the majority (80%) of patients. Although loss of histone H3 lysine 27 trimethylation (H3K27me3) has been shown during MCC tumorigenesis, epigenetic dysregulation has largely been overlooked. METHODS We conducted global DNA methylation profiling of clinically annotated MCC primary tumors, metastatic skin tumors, metastatic lymph node tumors, paired normal tissues, and two human MCC cell lines using the Illumina Infinium EPIC DNA methylation BeadArray platform. RESULTS Significant differential DNA methylation patterns across the genome are revealed between the four tissue types, as well as based on MCPyV status. Furthermore, 964 genes directly regulated by promoter or gene body DNA methylation were identified with high enrichment in neuro-related pathways. Finally, our findings suggest that loss of H3K27me3 occupancy in MCC is attributed to KDM6B and EZHIP overexpression as a consequence of promoter DNA hypomethylation. CONCLUSIONS We have demonstrated specific DNA methylation patterns for primary MCC tumors, metastatic MCCs, and adjacent-normal tissues. We have also identified DNA methylation markers that not only show potential diagnostic or prognostic utility in MCC management, but also correlate with MCC tumorigenesis, MCPyV expression, neuroendocrine features, and H3K27me3 status. The identification of DNA methylation alterations in MCC supports the need for further studies to understand the clinical implications of epigenetic dysregulation and potential therapeutic targets in MCC.
Collapse
Affiliation(s)
- Hemant Gujar
- Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| | - Arjun Mehta
- Department of Biochemistry and Molecular Medicine, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| | - Hong-Tao Li
- Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| | - Yvonne C. Tsai
- Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| | - Xiangning Qiu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Daniel J. Weisenberger
- Department of Biochemistry and Molecular Medicine, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| | - Miriam Galvonas Jasiulionis
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo 669 5 andar, Vila Clementino, São Paulo, SP 04039032 Brazil
| | - Gino K. In
- Department of Dermatology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| | - Gangning Liang
- Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
| |
Collapse
|
2
|
Giannetta E, La Salvia A, Rizza L, Muscogiuri G, Campione S, Pozza C, Colao AALI, Faggiano A. Are Markers of Systemic Inflammatory Response Useful in the Management of Patients With Neuroendocrine Neoplasms? Front Endocrinol (Lausanne) 2021; 12:672499. [PMID: 34367064 PMCID: PMC8339959 DOI: 10.3389/fendo.2021.672499] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Given the increasing incidence of neuroendocrine neoplasms (NENs) over the past few decades, a more comprehensive knowledge of their pathophysiological bases and the identification of innovative NEN biomarkers represents an urgent unmet need. There is still little advance in the early diagnosis and management of these tumors, due to the lack of sensible and specific markers with prognostic value and ability to early detect the response to treatment. Chronic systemic inflammation is a predisposing factor for multiple cancer hallmarks, as cancer proliferation, progression and immune-evading. Therefore, the relevance of inflammatory biomarkers has been identified as critical in several types of tumours, including NENs. A bidirectional relationship between chronic inflammation and development of NENs has been reported. Neuroendocrine cells can be over-stimulated by chronic inflammation, leading to hyperplasia and neoplastic transformation. As the modulation of inflammatory response represents a therapeutic target, inflammatory markers could represent a promising new key tool to be applied in the diagnosis, the prediction of response to treatment and also as prognostic biomarkers in NENs field. The present review provides an overview of the pre-clinical and clinical data relating the potentially usefulness of circulating inflammatory markers: neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), cytokines and tissue inflammatory markers (PD-1/PD-L1), in the management of NENs. (1) NLR and PLR have both demonstrated to be promising and simple to acquire biomarkers in patients with advanced cancer, including NEN. To date, in the context of NENs, the prognostic role of NLR and PLR has been confirmed in 15 and 4 studies, respectively. However, the threshold value, both for NLR and PLR, still remains not defined. (2) Cytokines seem to play a central role in NENs tumorigenesis. In particular, IL-8 levels seems to be a good predictive marker of response to anti-angiogenic treatments. (3) PD-1 and PD-L1 expression on tumour cells and on TILs, have demonstrated to be promising predictive and prognostic biomarkers in NENs. Unfortunately, these two markers have not been validated so far and further studies are needed to establish their indications and utility.
Collapse
Affiliation(s)
- Elisa Giannetta
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
- *Correspondence: Elisa Giannetta,
| | - Anna La Salvia
- Department of Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Laura Rizza
- Endocrinology Unit, Department of Oncology and Medical Specialities, AO San Camillo-Forlanini, Rome, Italy
| | - Giovanna Muscogiuri
- Endocrinology Unit Department of Clinical Medicine and Surgery, University Federico II School of Medicine, Naples, Italy
| | - Severo Campione
- A. Cardarelli Hospital, Naples Department of Advanced Diagnostic-Therapeutic Technologies and Health Services Section of Anatomic Pathology, Naples, Italy
| | - Carlotta Pozza
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | | | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, Endocrine-Metabolic Unit, Sant’Andrea University Hospital “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|
3
|
Chandra S, Zheng Y, Pandya S, Yu T, Kearney M, Wang L, Kim R, Phatak H. Real-world outcomes among US Merkel cell carcinoma patients initiating immune checkpoint inhibitors or chemotherapy. Future Oncol 2020; 16:2521-2536. [PMID: 32883109 DOI: 10.2217/fon-2020-0453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: Retrospectively assessed treatment patterns and clinical and economic outcomes in Merkel cell carcinoma (MCC) patients receiving recommended first-line regimens. Materials & methods: MCC patients newly treated with either immune checkpoint inhibitors (ICIs) or chemotherapies (CTs) were selected from the Veterans Health Administration database (2013-2018); 74 patients (ICIs: 20 and CTs: 54) were selected. Results: Median duration of therapy was 300 days for ICIs and 91 days for CTs. Time to next treatment was 245 and 184 days, respectively. Mean total (per patient per month) costs were $15,306 (ICIs) and $10,957 (CTs), of which 51% and 86%, respectively, were non-MCC therapy-related costs. Conclusion: Despite higher costs, utilization of ICIs in first-line MCC shows clinical advantages over CTs in the real world.
Collapse
Affiliation(s)
| | - Ying Zheng
- EMD Serono Inc., Rockland, MA 02370, USA
| | | | - Ting Yu
- EMD Serono Inc., Rockland, MA 02370, USA
| | | | - Li Wang
- STATinMED Research, Plano, TX 75024, USA
| | - Ruth Kim
- Pfizer Inc., New York, NY 10017, USA
| | | |
Collapse
|
4
|
Wu S, Slater NA, Sayed CJ, Googe PB. PD‐L1
and
LAG
‐3 expression in advanced cutaneous squamous cell carcinomas. J Cutan Pathol 2020; 47:882-887. [DOI: 10.1111/cup.13709] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 03/03/2020] [Accepted: 04/07/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Sam Wu
- Department of Dermatology University of North Carolina Chapel Hill North Carolina USA
| | - Nathaniel A. Slater
- Department of Dermatology University of North Carolina Chapel Hill North Carolina USA
| | - Christopher J. Sayed
- Department of Dermatology University of North Carolina Chapel Hill North Carolina USA
| | - Paul B. Googe
- Department of Dermatology University of North Carolina Chapel Hill North Carolina USA
| |
Collapse
|
5
|
Gortman A. Definitive Radiotherapy for Locally Advanced Merkel Cell Carcinoma of the Head and Neck Region: A Case Report. Cureus 2019; 11:e6270. [PMID: 31903306 PMCID: PMC6937466 DOI: 10.7759/cureus.6270] [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] [Indexed: 11/07/2022] Open
Abstract
Merkel cell carcinoma (MCC) is a highly aggressive neuroendocrine malignancy of skin origin. It is most commonly managed with upfront surgical resection which is then followed by radiotherapy as soon as possible postoperatively. Radiotherapy alone has been used in inoperable cases and in cases of patient preference to omit surgical management. MCC is a very radiosensitive disease, and an ideal location for definitive radiotherapy is the head and neck region (which represents the majority of cases), where resection often leads to reduced outcomes given the number of critical structures in the region and the inability of surgery to achieve the excision margins required for the aggressive nature of the disease without leading to severely impacted cosmesis and function. This case study looks at an elderly woman who was treated with definitive dose fractionation (to a total dose of 60 Gray in 30 fractions) radiotherapy to the head and neck region for Stage III, T1N2M0 disease, following rapid loco-regional relapse after surgical wide local excision. Two years postdefinitive radiotherapy, she has had complete loco-regional control both clinically and radiologically.
Collapse
Affiliation(s)
- Aron Gortman
- Radiation Oncology, Lismore Base Hospital, North Coast Cancer Institute, Lismore, AUS
| |
Collapse
|
6
|
Shibayama Y, Kameda H, Ota S, Tsuchida K, Cho KY, Nakamura A, Miyoshi H, Atsumi T. Case of fulminant type 1 diabetes induced by the anti-programmed death-ligand 1 antibody, avelumab. J Diabetes Investig 2019; 10:1385-1387. [PMID: 30738003 PMCID: PMC6717815 DOI: 10.1111/jdi.13022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/03/2019] [Accepted: 02/05/2019] [Indexed: 12/19/2022] Open
Abstract
With the expansive use of immune checkpoint inhibitors, the frequency of immune-related adverse events, including autoimmune type 1 diabetes, has been exponentially increased. The anti-programmed death-ligand 1 antibody, avelumab, has recently been approved for metastatic Merkel cell carcinoma therapy. Here, we report a patient that developed fulminant type 1 diabetes during avelumab treatment. An 81-year-old woman with no history of diabetes received avelumab for metastatic Merkel cell carcinoma. Elevated plasma glucose level (483 mg/dL), hemoglobin A1c level (7.5%) and ketosis were observed after 10 courses of avelumab without any symptoms related to hyperglycemia. As the laboratory tests showed insulin depletion, we diagnosed her with fulminant type 1 diabetes induced by avelumab. This is the first reported case of avelumab-induced type 1 diabetes, illustrating the necessity for close monitoring of glycemic control during avelumab therapy, as well as other immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yui Shibayama
- Department of Rheumatology, Endocrinology and NephrologyFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Hiraku Kameda
- Department of Rheumatology, Endocrinology and NephrologyFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Shoichiro Ota
- Postgraduate Clinical Training CenterHokkaido University HospitalSapporoJapan
| | - Kazuhisa Tsuchida
- Department of Rheumatology, Endocrinology and NephrologyFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Kyu Yong Cho
- Department of Rheumatology, Endocrinology and NephrologyFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Akinobu Nakamura
- Department of Rheumatology, Endocrinology and NephrologyFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Hideaki Miyoshi
- Division of Diabetes and ObesityFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and NephrologyFaculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| |
Collapse
|
7
|
Westbrook BC, Norwood TG, Terry NLJ, McKee SB, Conry RM. Talimogene laherparepvec induces durable response of regionally advanced Merkel cell carcinoma in 4 consecutive patients. JAAD Case Rep 2019; 5:782-786. [PMID: 31516997 PMCID: PMC6728723 DOI: 10.1016/j.jdcr.2019.06.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Key Words
- CR, complete response
- CT, computed tomography
- FDA, US Food and Drug Administration
- ICB, immune checkpoint blockade
- MCC, Merkel cell carcinoma
- MCPyV, Merkel cell polyomavirus
- Merkel cell carcinoma
- ORR, objective response rate
- PD-L1, programmed death ligand 1
- PET, positron emission tomography
- PFS, progression-free survival
- SUV, standardized uptake values
- TVEC, talimogene laherparepvec
- advanced Merkel cell carcinoma
- durable response
- immunotherapy
- oncolytic virus
- regionally advanced Merkel cell carcinoma
- talimogene laherparepvec
Collapse
Affiliation(s)
- Brian C Westbrook
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - T Graham Norwood
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Nina L J Terry
- Department of Radiology, General Radiology and Cardiopulmonary Section, University of Alabama at Birmingham Medicine, Birmingham, Alabama
| | - Svetlana B McKee
- Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert M Conry
- Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
8
|
Villani A, Fabbrocini G, Costa C, Carmela Annunziata M, Scalvenzi M. Merkel Cell Carcinoma: Therapeutic Update and Emerging Therapies. Dermatol Ther (Heidelb) 2019; 9:209-222. [PMID: 30820877 PMCID: PMC6522614 DOI: 10.1007/s13555-019-0288-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Indexed: 02/08/2023] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but highly aggressive neuroendocrine skin cancer whose incidence has almost doubled in recent decades. Risk factors for MCC include age > 65 years, immunosuppression, sun exposure and infection by Merkel cell polyomavirus. MCC usually presents as rapidly growing, firm, red to violaceous nodule localized on the sun-exposed skin. Surgery followed by radiation therapy is considered to be the first-line treatment for primary or loco-regional MCC in order to prevent recurrences and lymph node metastasis, while chemotherapy has always been used to treat advanced forms. However, responses to chemotherapy are mostly of short duration, and the associated clinical benefit on overall survival is still unclear. The use of checkpoint inhibitors (CPIs) has shown good results in the treatment of advanced MCC and, consequently, CPIs are considered emerging immunotherapeutic options for these patients, although there are still no standardized treatments for patients with metastatic disease. Here we present a complete overview of the different possibilities for the treatment of MCC according to the stage of the disease, focusing on the emerging immunotherapies used for treating advanced MCC.
Collapse
Affiliation(s)
- Alessia Villani
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico, Naples, Italy.
| | - Gabriella Fabbrocini
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico, Naples, Italy
| | - Claudia Costa
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico, Naples, Italy
| | - Maria Carmela Annunziata
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico, Naples, Italy
| | - Massimiliano Scalvenzi
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico, Naples, Italy
| |
Collapse
|
9
|
Looi CK, Chung FFL, Leong CO, Wong SF, Rosli R, Mai CW. Therapeutic challenges and current immunomodulatory strategies in targeting the immunosuppressive pancreatic tumor microenvironment. J Exp Clin Cancer Res 2019; 38:162. [PMID: 30987642 PMCID: PMC6463646 DOI: 10.1186/s13046-019-1153-8] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/22/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal type of cancers, with an overall five-year survival rate of less than 5%. It is usually diagnosed at an advanced stage with limited therapeutic options. To date, no effective treatment options have demonstrated long-term benefits in advanced pancreatic cancer patients. Compared with other cancers, pancreatic cancer exhibits remarkable resistance to conventional therapy and possesses a highly immunosuppressive tumor microenvironment (TME). MAIN BODY In this review, we summarized the evidence and unique properties of TME in pancreatic cancer that may contribute to its resistance towards immunotherapies as well as strategies to overcome those barriers. We reviewed the current strategies and future perspectives of combination therapies that (1) promote T cell priming through tumor associated antigen presentation; (2) inhibit tumor immunosuppressive environment; and (3) break-down the desmoplastic barrier which improves tumor infiltrating lymphocytes entry into the TME. CONCLUSIONS It is imperative for clinicians and scientists to understand tumor immunology, identify novel biomarkers, and optimize the position of immunotherapy in therapeutic sequence, in order to improve pancreatic cancer clinical trial outcomes. Our collaborative efforts in targeting pancreatic TME will be the mainstay of achieving better clinical prognosis among pancreatic cancer patients. Ultimately, pancreatic cancer will be a treatable medical condition instead of a death sentence for a patient.
Collapse
Affiliation(s)
- Chin-King Looi
- 0000 0000 8946 5787grid.411729.8School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Felicia Fei-Lei Chung
- Mechanisms of Carcinogenesis Section (MCA), Epigenetics Group (EGE) International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Chee-Onn Leong
- 0000 0000 8946 5787grid.411729.8School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- 0000 0000 8946 5787grid.411729.8Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| | - Shew-Fung Wong
- 0000 0000 8946 5787grid.411729.8School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Rozita Rosli
- 0000 0001 2231 800Xgrid.11142.37UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Sri Kembangan, Selangor Malaysia
| | - Chun-Wai Mai
- 0000 0000 8946 5787grid.411729.8School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- 0000 0000 8946 5787grid.411729.8Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
10
|
Patel K, Siraj S, Smith C, Nair M, Vishwanatha JK, Basha R. Pancreatic Cancer: An Emphasis on Current Perspectives in Immunotherapy. Crit Rev Oncog 2019; 24:105-118. [PMID: 31679206 PMCID: PMC8038975 DOI: 10.1615/critrevoncog.2019031417] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pancreatic cancer affects both male and female individuals with higher incidences and death rates among the male population. Detection of this malignancy is delayed due to the lack of symptoms in the early-stage cancer, which makes it extremely difficult to treat. Identifying effective strategies has been a challenge for improving the survival rates in pancreatic cancer patients. Resistance to chemotherapy is often developed in pancreatic cancer treatment. Although many strategies are under clinical trials to target certain markers associated with cancer, immunotherapeutic approaches are currently gaining importance. Immunotherapy for pancreatic cancer is in the limelight after preclinical research showed some promise. Immunotherapy approaches were tested along with other treatment options to enhance the treatment effect. Adoptive cell transfer and immune checkpoint inhibitors are currently in clinical trials. The Food and Drug Administration approved pembrolizumab in a fast-tracked review for advanced pancreatic cancer patients. Pembrolizumab blocks the checkpoint protein, programmed cell death protein 1 (PD-1), on T cells to boost the response of the immune system against cancer cells, thereby shrinking tumors. The recent developments in immunotherapy and the early success in other cancers are encouraging to further test immunotherapy in pancreatic cancer. The combination of pembrolizumab and pelareorep, an isolate of human reovirus, is in phase II clinical study in metastatic disease. Depending on the results of current clinical trials and testing, the strategies in the pipeline are expected to increase the use of immunotherapy in the clinical testing setting. Success in immunotherapy is urgently needed to address the side-effects, treating patients with advanced disease and reducing metastasis for increasing the survival rate in pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | - Chloe Smith
- Old Dominion University, Norfolk, Virginia 23529
| | - Maya Nair
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, Texas 76107
| | - Jamboor K. Vishwanatha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, Texas 76107
| | | |
Collapse
|
11
|
Rudzki JD. Management of adverse events related to checkpoint inhibition therapy. MEMO 2018; 11:132-137. [PMID: 29983828 PMCID: PMC6006276 DOI: 10.1007/s12254-018-0416-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/17/2018] [Indexed: 12/17/2022]
Abstract
IO treatments (immuno-oncology treatments) have become reality and are now daily practice or, in some cases, a daily challenge. New recommendations are being made with the prime purpose of increasing alertness and awareness as well as emphasizing standard operating strategies to deal with immune-related adverse events (ir-AEs) in patients treated with immune checkpoint inhibitors (ICI). This brief review refers to systemic reviews, guidelines and meta-analyses, randomized controlled trials and case series published from 2000 to the present. Existing recommendations for optimal management of toxicities vary according to organ systems affected and grading. Grade 1 toxicities (exception to the rule: neurologic, hematologic, cardiac manifestation) require close monitoring. Grade 2 toxicities prompt immediate treatment interruption combined with corticosteroid administration (prednisone or methylprednisolone 0.5-1 mg/kg/day) until the symptoms revert to grade 1 or less. ir-AEs up to grade 3 or 4 justify suspension of treatment together with increased dosage of prednisone or methylprednisolone (1-2 mg/kg/day) combined with close monitoring to continuously adapt the current immunosuppressive strategy. In some cases, a different additional immunosuppressive agent has to be evaluated. Only when all symptoms have disappeared and immunosuppressive treatment produces a response can all immunosuppressive agents be tapered. Endocrinopathies are the exception to the rule and are mostly controllable by hormone replacement, at least in low-grade manifestation. This short review focuses on the main aspects that help manage immune-related side-effects and elucidates all the additional aspects surrounding and contributing to successful treatment and management of cancer patients.
Collapse
Affiliation(s)
- Jakob Daniel Rudzki
- Medical University Innsbruck, Innsbruck, Austria
- UKIM V—Tirol Kliniken, Landeskrankenhaus Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| |
Collapse
|