1
|
Mitsunaga K, Bagot M, Ram-Wolff C, Guenova E, von Gugelberg C, Hodak E, Amitay-Laish I, Papadavid E, Jonak C, Porkert S, Scarisbrick J, Applewaite R, Beylot-Barry M, Nicolay J, Quaglino P, Sanches JA, Cury-Martins J, Lora-Pablos D, Ortiz P. Real-world study of pegylated interferon α-2a to treat mycosis fungoides/Sézary syndrome using time to next treatment as a measure of clinical benefit: an EORTC CLTG study. Br J Dermatol 2024; 191:419-427. [PMID: 38596857 DOI: 10.1093/bjd/ljae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Mycosis fungoides (MF) and Sézary syndrome (SS) are chronic malignant diseases that typically necessitate diverse strategies to achieve remission. Systemic interferon (IFN)-α (subtypes 2a and 2b) has been used to treat MF/SS since 1984; however, its production was recently stopped. The recombinant pegylated (PEG) form of IFN-α-2a remains the only alternative IFN treatment, although it has not been approved for use in MF/SS. OBJECTIVES To assess the effectiveness and safety of PEG-IFN-α-2a in monotherapy and in combination with other treatments using time to next treatment (TTNT) as a measure of clinical therapeutic benefit in a real-world setting. METHODS We conducted an international, multicentre retrospective study of patients with MF and SS (of any stage) treated with PEG-IFN-α-2a from July 2012 to February 2022. Patients were included across 11 centres in 10 countries. The primary endpoints were to determine the TTNT of PEG-IFN-α-2a and adverse events (AEs) in MF/SS. RESULTS In total, 105 patients were included [mean (SD) age 61 (13.1) years]; 42 (40.0%) had stage IA-IIA and 63 (60.0%) had stage IIB-IVB disease. PEG-IFN-α-2a was combined with other therapies in 67 (63.8%) patients, most commonly with extracorporeal photopheresis (36%) and bexarotene (22%). Patients with stage I-IIA disease achieved an overall response rate (ORR) of 57%; the ORR in those with stage IIB-IVB disease was 51%. Combination treatment resulted in a median TTNT of 10.4 months (range 0.6-50.7) vs. 7.0 months (range 0.7-52.4) for those who received monotherapy (P < 0.01). Overall, the mean (SD) TTNT was 9.2 (10.6) months and the ORR was 53.3% (n = 56). A complete response was seen in 13% of patients and a partial response in 40%. AEs were described in 68.6% (n = 72) of patients. Flu-like symptoms (n = 28; 26.7%), lymphopenia (n = 24; 22.9%) and elevated liver function (n = 10; 9.5%) were the most frequently reported. Grade 3-4 AEs were reported in 23 (21.9%) patients, mostly related to myelosuppression. CONCLUSIONS PEG-IFN-α-2a for MF/SS resulted in an ORR of 53.3% and a mean (SD) TTNT of 9.2 (10.6) months. Combination regimens were superior to monotherapy and doses of 180 µg PEG-IFN-α-2a weekly were related to a higher ORR.
Collapse
Affiliation(s)
- Keila Mitsunaga
- Department of Dermatology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Martine Bagot
- Department of Dermatology, Université Paris Cité, Saint-Louis Hospital, Paris, France
| | - Caroline Ram-Wolff
- Department of Dermatology, Université Paris Cité, Saint-Louis Hospital, Paris, France
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital Zurich and Faculty of Medicine, Zurich, Switzerland
- Department of Dermatology, Lausanne University Hospital (CHUV) and Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Christina von Gugelberg
- Department of Dermatology, University Hospital Zurich and Faculty of Medicine, Zurich, Switzerland
- Department of Dermatology, Lausanne University Hospital (CHUV) and Faculty of Biology and Medicine, Lausanne, Switzerland
| | - Emmilia Hodak
- Division of Dermatology, Rabin Medical Center - Beilinson Hospital, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Amitay-Laish
- Division of Dermatology, Rabin Medical Center - Beilinson Hospital, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Evangelia Papadavid
- 2nd Department of Dermatology and Venereology, National and Kapodistrian University of Athens, 'Attikon' University General Hospital, Athens, Greece
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Stefanie Porkert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | | | - Marie Beylot-Barry
- Department of Dermatology, Bordeaux University Hospital Center, Bordeaux, France
| | - Jan Nicolay
- Department of Dermatology, Universitätsmedizin Mannheim, Mannheim, Germany
| | - Pietro Quaglino
- Department of Medical Science, University of Turin Medical School, Turin, Italy
| | | | | | - David Lora-Pablos
- Scientific Support Unit (i+12), Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pablo Ortiz
- Department of Dermatology, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
2
|
Grandi V, Colantuono VA, Pimpinelli N. Skin-directed therapy and biologic response modifiers in mycosis fungoides. Dermatol Reports 2024; 16:9926. [PMID: 39295879 PMCID: PMC11406212 DOI: 10.4081/dr.2024.9926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 09/21/2024] Open
Abstract
The most common and widespread type of cutaneous T-cell lymphoma is mycosis fungoides (MF), and it has a multiphasic clinical and biological course, with early stages being indolent for many years and later stages being faster and more aggressive. The clinical stage has a significant impact on the management and course of treatment: in the early stages, skin-directed therapies (SDT) plus/or biologic response modifiers (BRM); in the later stages, radiotherapy and/or systemic therapies. Even though national and international societies and groups periodically update their clinical recommendations, there is still no universally accepted approach. This paper reviews and discusses the various SDT and BRM options, either separately or in combination.
Collapse
Affiliation(s)
- Vieri Grandi
- Dermatology Unit, Department of Health Sciences, University of Florence Medical School, Florence; Melanoma and Skin Cancer Unit, Central Tuscany Health District, Italy
| | - Virginia Alba Colantuono
- Dermatology Unit, Department of Health Sciences, University of Florence Medical School, Florence; Melanoma and Skin Cancer Unit, Central Tuscany Health District, Italy
| | - Nicola Pimpinelli
- Dermatology Unit, Department of Health Sciences, University of Florence Medical School, Florence; Melanoma and Skin Cancer Unit, Central Tuscany Health District, Italy
| |
Collapse
|
3
|
Guglielmo A, Zengarini C, Agostinelli C, Motta G, Sabattini E, Pileri A. The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets. Cells 2024; 13:584. [PMID: 38607023 PMCID: PMC11012008 DOI: 10.3390/cells13070584] [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: 12/06/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Cutaneous T cell lymphomas (CTCLs), encompassing mycosis fungoides (MF) and Sézary syndrome (SS), present a complex landscape influenced by cytokines and cellular responses. In this work, the intricate relationship between these inflammatory proteins and disease pathogenesis is examined, focusing on what is known at the clinical and therapeutic levels regarding the most well-known inflammatory mediators. An in-depth look is given to their possible alterations caused by novel immunomodulatory drugs and how they may alter disease progression. From this narrative review of the actual scientific landscape, Interferon-gamma (IFN-γ) emerges as a central player, demonstrating a dual role in both promoting and inhibiting cancer immunity, but the work navigates through all the major interleukins known in inflammatory environments. Immunotherapeutic perspectives are elucidated, highlighting the crucial role of the cutaneous microenvironment in shaping dysfunctional cell trafficking, antitumor immunity, and angiogenesis in MF, showcasing advancements in understanding and targeting the immune phenotype in CTCL. In summary, this manuscript aims to comprehensively explore the multifaceted aspects of CTCL, from the immunopathogenesis and cytokine dynamics centred around TNF-α and IFN-γ to evolving therapeutic modalities. Including all the major known and studied cytokines in this analysis broadens our understanding of the intricate interplay influencing CTCL, paving the way for improved management of this complex lymphoma.
Collapse
Affiliation(s)
- Alba Guglielmo
- Institute of Dermatology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
| | - Corrado Zengarini
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Claudio Agostinelli
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giovanna Motta
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Elena Sabattini
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Alessandro Pileri
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
4
|
Goel RR, Rook AH. Psoralen Plus UVA Induces Local IFN Production and Antitumor Responses in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2024; 144:449-450. [PMID: 37921716 DOI: 10.1016/j.jid.2023.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 11/04/2023]
Affiliation(s)
- Rishi R Goel
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Institute for Immunology and Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alain H Rook
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
5
|
Yu Z, Vieyra-Garcia P, Benezeder T, Crouch JD, Kim IR, O'Malley JT, Devlin PM, Gehad A, Zhan Q, Gudjonsson JE, Sarkar MK, Kahlenberg JM, Gerard N, Teague JE, Kupper TS, LeBoeuf NR, Larocca C, Tawa M, Pomahac B, Talbot SG, Orgill DP, Wolf P, Clark RA. Phototherapy Restores Deficient Type I IFN Production and Enhances Antitumor Responses in Mycosis Fungoides. J Invest Dermatol 2024; 144:621-632.e1. [PMID: 37716650 PMCID: PMC10922223 DOI: 10.1016/j.jid.2023.06.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 09/18/2023]
Abstract
Transcriptional profiling demonstrated markedly reduced type I IFN gene expression in untreated mycosis fungoides (MF) skin lesions compared with that in healthy skin. Type I IFN expression in MF correlated with antigen-presenting cell-associated IRF5 before psoralen plus UVA therapy and epithelial ULBP2 after therapy, suggesting an enhancement of epithelial type I IFN. Immunostains confirmed reduced baseline type I IFN production in MF and increased levels after psoralen plus UVA treatment in responding patients. Effective tumor clearance was associated with increased type I IFN expression, enhanced recruitment of CD8+ T cells into skin lesions, and expression of genes associated with antigen-specific T-cell activation. IFNk, a keratinocyte-derived inducer of type I IFNs, was increased by psoralen plus UVA therapy and expression correlated with upregulation of other type I IFNs. In vitro, deletion of keratinocyte IFNk decreased baseline and UVA-induced expression of type I IFN and IFN response genes. In summary, we find a baseline deficit in type I IFN production in MF that is restored by psoralen plus UVA therapy and correlates with enhanced antitumor responses. This may explain why MF generally develops in sun-protected skin and suggests that drugs that increase epithelial type I IFNs, including topical MEK and EGFR inhibitors, may be effective therapies for MF.
Collapse
Affiliation(s)
- Zizi Yu
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pablo Vieyra-Garcia
- Research Unit for Photodermatology, Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Theresa Benezeder
- Research Unit for Photodermatology, Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Jack D Crouch
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ira R Kim
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John T O'Malley
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Phillip M Devlin
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmed Gehad
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qian Zhan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nega Gerard
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica E Teague
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas S Kupper
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Center for Cutaneous Oncology, Dana-Farber Cancer Institute/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicole R LeBoeuf
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Center for Cutaneous Oncology, Dana-Farber Cancer Institute/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Cecilia Larocca
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Center for Cutaneous Oncology, Dana-Farber Cancer Institute/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianne Tawa
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Bohdan Pomahac
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Simon G Talbot
- Division of Plastic and Reconstructive Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dennis P Orgill
- Division of Plastic and Reconstructive Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Wolf
- Research Unit for Photodermatology, Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria.
| | - Rachael A Clark
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
6
|
Aires D, Abhyankar S. Early intervention of extracorporeal photopheresis for advancing/progressing cutaneous T-cell lymphoma. Hematol Oncol 2023; 41:809-816. [PMID: 37974524 DOI: 10.1002/hon.3229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 08/18/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023]
Abstract
Patients with cutaneous T-cell lymphoma with progressive disease typically undergo a series of skin-directed and systemic therapy regimens during cycles of response and relapse. Extracorporeal photopheresis (ECP) is an effective and safe systemic treatment option, often reserved for later stages of disease and typically employed after failure of several other therapies. ECP has benefits in response rate, time to next treatment, and tolerability that may support its use earlier in the treatment cycle for advancing/progressing disease.
Collapse
Affiliation(s)
| | - Sunil Abhyankar
- University of Kansas Cancer Center and the University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
7
|
Fahmy LM, Schreidah CM, Lapolla BA, Magro CM, Geskin LJ. Mycosis fungoides diagnosed after exposure to risankizumab for psoriasis. JAAD Case Rep 2023; 41:85-89. [PMID: 37916040 PMCID: PMC10615897 DOI: 10.1016/j.jdcr.2023.08.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Affiliation(s)
- Lauren M. Fahmy
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| | - Celine M. Schreidah
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| | - Brigit A. Lapolla
- Department of Dermatology, Columbia University Irving Medical Center, New York, New York
| | - Cynthia M. Magro
- Department of Pathology and Laboratory Medicine, Division of Dermatopathology, Weill Cornell Medicine, New York, New York
| | - Larisa J. Geskin
- Department of Dermatology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
8
|
Gosmann J, Stadler R, Quint KD, Gutzmer R, Vermeer MH. Use of Pegylated Interferon Alpha-2a in Cutaneous T-cell Lymphoma: A Retrospective Case Collection. Acta Derm Venereol 2023; 103:adv10306. [PMID: 37902466 PMCID: PMC10622159 DOI: 10.2340/actadv.v103.10306] [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: 02/28/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Mycosis fungoides and Sézary syndrome are rare and largely incurable types of cutaneous T-cell lymphoma with limited therapeutic options. In 1984 Bunn et al. reported that interferon alpha is an efficient monotherapy in cutaneous T-cell lymphoma and 14 years later it was shown in a prospective, randomized trial that a combination of interferon alpha and psoralen plus ultraviolet A therapy (PUVA) is most efficient in the treatment of cutaneous T-cell lymphoma. Since then interferon alpha as single agent or, most often, in combination with phototherapy and/or retinoids has been integrated as standard of care in cutaneous T-cell lymphoma guidelines worldwide. However, production of interferon alpha was discontinued recently worldwide and pegylated interferon alpha-2a (PEG-IFNα) has been used as an alternative therapy. In contrast to numerous interferon alpha studies, only a few studies focusing on PEG-IFNα are available. Therefore, the aim of this study was to conduct a retrospective data collection to report on the efficacy, adverse events and therapy regimens of PEG-IFNα in cutaneous T-cell lymphoma. In 28 patients with cutaneous T-cell lymphoma treated in Germany and in the Netherlands, 36% of patients achieved complete remission, 36% partial remission and 29% stable disease. Eighteen percent of patients developed adverse events during therapy, which led to the discontinuation of PEG-IFNα therapy in 2 patients. The most common concomittant therapies were oral PUVA phototherapy and local radiotherapy. In conclusion, PEG-IFNα, especially in combination with skin-directed therapies, is an effective treatment option for cutaneous T-cell lymphoma in clinical practice.
Collapse
Affiliation(s)
- Janika Gosmann
- University Department for Dermatology, Venereology, Allergology and Phlebology Skin Cancer Center, Johannes Wesling Medical Center Minden, Ruhr University Bochum, Minden, Germany
| | - Rudolf Stadler
- University Department for Dermatology, Venereology, Allergology and Phlebology, Skin Cancer Center, Johannes Wesling Medical Center Minden, Ruhr University Bochum, Minden, Germany.
| | - Koen D Quint
- Department of Dermatology of the Leiden University medical Center (LUMC), Leiden, the Netherlands
| | - Ralf Gutzmer
- University Department for Dermatology, Venereology, Allergology and Phlebology Skin Cancer Center, Johannes Wesling Medical Center Minden, Ruhr University Bochum, Minden, Germany
| | - Maarten H Vermeer
- Department of Dermatology of the Leiden University medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
9
|
Hoshino T, Murayama N, Yamagishi K, Okado Y, Iwai H, Shirai K, Hosaka S, Moore PF, Nagata M. Clinical efficacy of recombinant canine interferon-gamma therapy in dogs with cutaneous epitheliotropic T-cell lymphoma. Vet Dermatol 2023; 34:460-467. [PMID: 37006127 DOI: 10.1111/vde.13161] [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: 12/22/2021] [Revised: 12/20/2022] [Accepted: 02/19/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND The antitumour effects of interferon-gamma (IFN-γ) in humans with cutaneous epitheliotropic T-cell lymphoma (CETCL) have been described; however, the efficacy of IFN-γ in dogs has not been investigated. HYPOTHESIS/OBJECTIVES The aim of this study was to evaluate the efficacy of recombinant canine IFN-γ (rCaIFN-γ) therapy in dogs with CETCL. ANIMALS Twenty dogs with CETCL recruited from seven veterinary clinics were enrolled in the study. MATERIALS AND METHODS Fifteen dogs were treated with rCaIFN-γ, and five control dogs were treated with prednisolone. We evaluated survival time, skin lesions (erythema, nodules, ulcers and bleeding), pruritus and general condition (sleep, appetite and body weight). In the rCaIFN-γ group, a questionnaire regarding the therapy was administered to owners after the dogs died. RESULTS No significant differences existed in the median survival time between the rCaIFN-γ and control groups (log-rank test: p = 0.2761, Wilcoxon's rank sum test: p = 0.4444). However, there were significant differences in ulcer, bleeding, pruritus, sleep, appetite and body weight between the groups (Wilcoxon-Mann-Whitney U-test: p = 0.0023, p = 0.0058, p = 0.0005, p = 0.0191, p = 0.0306 and p = 0.0306, respectively). Two (40%) of five dogs were euthanised in the control group, compared with none in the rCaIFN-γ group. Fourteen questionnaires were collected, and owners reported that they were satisfied with the rCaIFN-γ treatment. CONCLUSIONS AND CLINICAL RELEVANCE Although the median survival time was not prolonged, rCaIFN-γ could be helpful in maintaining good quality of life for dogs with CETCL.
Collapse
Affiliation(s)
- Tomoya Hoshino
- Department of Dermatology, Veterinary Specialists Emergency Centre, Saitama, Japan
| | | | | | | | | | | | | | - Peter F Moore
- Department of Veterinary Pathology, University of California, Davis, California, USA
| | - Masahiko Nagata
- Department of Dermatology, Veterinary Specialists Emergency Centre, Saitama, Japan
- Dermatology Services for Dogs and Cats, Tokyo, Japan
- ASC, Tokyo, Japan
| |
Collapse
|
10
|
Hu ZH, Lu L, Feng JD, Song HB, Zhang SY, Yang L, Wang T, Liu YH. Real-World Clinical Characteristics, Management, and Outcomes of 44 Paediatric Patients with Hypopigmented Mycosis Fungoides. Acta Derm Venereol 2023; 103:adv6226. [PMID: 37606155 PMCID: PMC10461309 DOI: 10.2340/actadv.v103.6226] [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: 12/14/2022] [Accepted: 06/19/2023] [Indexed: 08/23/2023] Open
Abstract
Hypopigmented mycosis fungoides is a rare form of mycosis fungoides that is characterized by achromic lesions, early onset of disease, a predilection for darker skinned populations, and a predominance of CD8+ T cells. Due to the rarity and heterogeneous presentation of hypopigmented mycosis fungoides, there are no criteria that clearly define the clinical characteristics and treatment regimens for this condition. This retrospective study of 44 paediatric patients with hypopigmented mycosis fungoides aimed to summarize their epidemiological and clinical characteristics and assess the effectiveness and safety of different treatment regimens. Clinical manifestations were further classified into 3 morphological groups: hypopigmented lesions, papules overlying hypopigmented lesions, and erythematous plaques overlying hypopigmented lesions. In addition, the results of this study suggest that interferon alpha might be an effective and well-tolerated therapy that could shorten the treatment time to complete response compared with other treatments. Maintenance therapy and long-term follow-up reduced the recurrence rate.
Collapse
Affiliation(s)
- Zhong-Hui Hu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Lu Lu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Jin-Di Feng
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Hong-Bin Song
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Shi-Yu Zhang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Lu Yang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Tao Wang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| | - Yue-Hua Liu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| |
Collapse
|
11
|
Transitioning to Pegylated Interferon for the Treatment of Cutaneous T-Cell Lymphoma: Meeting the Challenge of Therapy Discontinuation and a Proposed Algorithm. Dermatol Ther 2023. [DOI: 10.1155/2023/7171937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Cutaneous T-cell lymphoma (CTCL) is an uncommon non-Hodgkin lymphoma characterized by skin involvement, with the most recognized subtypes being mycosis fungoides (MF) and Sezary syndrome (SS). Interferon has been an established treatment for MF/SS since 1984 and is integrated into management guidelines internationally. In 2019, manufacturers abruptly discontinued interferon-α2b and interferon-α2a. Many alternative systemic therapies in MF/SS remain unfunded or unavailable in Canada, presenting a unique challenge. Although off-label use of pegylated interferon is a logical substitute, there are no established dosing guidelines and limited published experience. This case series provides a single-center experience on pegylated interferon-α2b for treatment of MF/SS, a suggested management algorithm, and a review of the literature. All patients identified in the Calgary Cutaneous Lymphoma Program with stage IIB–IVB MF/SS treated with interferon-α2b (4.5–9 MU/week) were switched to once weekly pegylated interferon (90 μg, 0.5 mL) between February and July 2021. Response was monitored using the mSWAT and SkinDex-29 tools. Eight patients were switched to pegylated interferon, with a median disease duration of 69 months (range: 8–275 months). Five out of eight patients remain on pegylated interferon, with the remainder having switched to preplanned therapies. Two patients required dose reduction due to side effects, including grade II anemia and mood changes. The remaining patients had normal laboratory investigations and no additional side effects. Uncommon lymphomas like MF/SS have limited treatment options, and the impact of abrupt product discontinuation is substantial. We propose a management algorithm for the transition of patients from interferon to pegylated interferon.
Collapse
|
12
|
Vorontsova AA, Karamova AE. Phototherapy combined with systemic agents for mycosis fungoides. VESTNIK DERMATOLOGII I VENEROLOGII 2022. [DOI: 10.25208/vdv1389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The choice of tactics for managing a patient with mycosis fungoides is determined by the stage of the disease. In the early stages of mycosis fungoides, a treatment approach using external therapy and various ultraviolet irradiation spectra (UVB-311 nm and PUVA therapy) is preferable. With insufficient efficiency and / or short remissions in some patients, it is possible to use combined methods in the early stages: PUVA therapy or UVB-311 nm therapy. in combination with systemic therapy, including interferon (IFN) 2b preparations, methotrexate and retinoids. The results of original studies demonstrate an increase in the overall response rate when combining PUVA therapy with IFN- or retinoids in patients with stages IB-IIB. The effectiveness of combined treatment regimens using UVB-311 nm remains insufficiently studied.
Collapse
|
13
|
Chen L, Deng J. Role of non-coding RNA in immune microenvironment and anticancer therapy of gastric cancer. J Mol Med (Berl) 2022; 100:1703-1719. [PMID: 36329206 DOI: 10.1007/s00109-022-02264-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Gastric cancer remains one of the cancers with the highest mortality in the world; therefore, it is very important to investigate its pathogenesis to improve the prognosis of gastric cancer patients. Recently, noncoding RNAs have become a research hotspot in the field of oncology. These RNA molecules play complex roles in the regulation of tumor cells, immune cells, and the tumor microenvironment. Therefore, studying their ability to regulate the gastric cancer immune microenvironment will provide us with a better perspective to understand their potential role in anticancer therapy. In this review, we discuss the regulatory effects of several common noncoding RNAs on the immune microenvironment of gastric cancer and their prospects in anticancer therapy to provide some novel insight into the identification of valuable diagnostic markers and improving the prognosis of gastric cancer patients.
Collapse
Affiliation(s)
- Liqiao Chen
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Jingyu Deng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
14
|
Micellar Curcumin Substantially Increases the Antineoplastic Activity of the Alkylphosphocholine Erufosine against TWIST1 Positive Cutaneous T Cell Lymphoma Cell Lines. Pharmaceutics 2022; 14:pharmaceutics14122688. [PMID: 36559182 PMCID: PMC9781439 DOI: 10.3390/pharmaceutics14122688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a rare form of cancer with local as well as systemic manifestations. Concomitant bacterial infections increase morbidity and mortality rates due to impaired skin barrier and immune deficiency. In the current study, we demonstrated that the in vitro anti-lymphoma potential of erufosine is diminished by TWIST1 expression and micellar curcumin substantially increases its antineoplastic activity. Pharmacokinetic analysis showed that the micellar curcumin (MCRM) used in our study was characterized by low zeta potential, slow release of curcumin, and fast cell membrane penetration. The combination ratio 1:4 [erufosine:MCRM] achieved strong synergism by inhibiting cell proliferation and clonogenicity. The combined antiproliferative effects were calculated using the symbolic mathematical software MAPLE 15. The synergistic combination strongly decreased the expression of TWIST1 and protein kinase B/Akt as proven by western blotting. Significant reductions in NF-κB activation, induction of apoptosis, and altered glutathione levels were demonstrated by corresponding assays. In addition, the synergistic combination enhanced the anti-staphylococcal activity and prevented biofilm formation, as shown by crystal violet staining. Taken together, the above results show that the development of nanotechnological treatment modalities for CTCL, based on rational drug combinations exhibiting parallel antineoplastic and antibacterial effects, may prove efficacious.
Collapse
|
15
|
Multi-Omics Approaches for the Prediction of Clinical Endpoints after Immunotherapy in Non-Small Cell Lung Cancer: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061237. [PMID: 35740259 PMCID: PMC9219996 DOI: 10.3390/biomedicines10061237] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the management of locally advanced and advanced non-small lung cancer (NSCLC). With an improvement in the overall survival (OS) as both first- and second-line treatments, ICIs, and especially programmed-death 1 (PD-1) and programmed-death ligands 1 (PD-L1), changed the landscape of thoracic oncology. The PD-L1 level of expression is commonly accepted as the most used biomarker, with both prognostic and predictive values. However, even in a low expression level of PD-L1, response rates remain significant while a significant number of patients will experience hyperprogression or adverse events. The dentification of such subtypes is thus of paramount importance. While several studies focused mainly on the prediction of the PD-L1 expression status, others aimed directly at the development of prediction/prognostic models. The response to ICIs depends on a complex physiopathological cascade, intricating multiple mechanisms from the molecular to the macroscopic level. With the high-throughput extraction of features, omics approaches aim for the most comprehensive assessment of each patient. In this article, we will review the place of the different biomarkers (clinical, biological, genomics, transcriptomics, proteomics and radiomics), their clinical implementation and discuss the most recent trends projecting on the future steps in prediction modeling in NSCLC patients treated with ICI.
Collapse
|
16
|
Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y, Xia Y. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol 2022; 15:24. [PMID: 35279217 PMCID: PMC8917703 DOI: 10.1186/s13045-022-01242-2] [Citation(s) in RCA: 186] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint molecules are promising anticancer targets, among which therapeutic antibodies targeting the PD-1/PD-L1 pathway have been widely applied to cancer treatment in clinical practice and have great potential. However, this treatment is greatly limited by its low response rates in certain cancers, lack of known biomarkers, immune-related toxicity, innate and acquired drug resistance, etc. Overcoming these limitations would significantly expand the anticancer applications of PD-1/PD-L1 blockade and improve the response rate and survival time of cancer patients. In the present review, we first illustrate the biological mechanisms of the PD-1/PD-L1 immune checkpoints and their role in the healthy immune system as well as in the tumor microenvironment (TME). The PD-1/PD-L1 pathway inhibits the anticancer effect of T cells in the TME, which in turn regulates the expression levels of PD-1 and PD-L1 through multiple mechanisms. Several strategies have been proposed to solve the limitations of anti-PD-1/PD-L1 treatment, including combination therapy with other standard treatments, such as chemotherapy, radiotherapy, targeted therapy, anti-angiogenic therapy, other immunotherapies and even diet control. Downregulation of PD-L1 expression in the TME via pharmacological or gene regulation methods improves the efficacy of anti-PD-1/PD-L1 treatment. Surprisingly, recent preclinical studies have shown that upregulation of PD-L1 in the TME also improves the response and efficacy of immune checkpoint blockade. Immunotherapy is a promising anticancer strategy that provides novel insight into clinical applications. This review aims to guide the development of more effective and less toxic anti-PD-1/PD-L1 immunotherapies.
Collapse
Affiliation(s)
- Mengling Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qianrui Huang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yao Xie
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, 610041, China
| | - Hongbo Ma
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Xia
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, 610041, China.
| |
Collapse
|
17
|
Gill RPK, Gantchev J, Martínez Villarreal A, Ramchatesingh B, Netchiporouk E, Akilov OE, Ødum N, Gniadecki R, Koralov SB, Litvinov IV. Understanding Cell Lines, Patient-Derived Xenograft and Genetically Engineered Mouse Models Used to Study Cutaneous T-Cell Lymphoma. Cells 2022; 11:cells11040593. [PMID: 35203244 PMCID: PMC8870189 DOI: 10.3390/cells11040593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Cutaneous T cell lymphoma (CTCL) is a spectrum of lymphoproliferative disorders caused by the infiltration of malignant T cells into the skin. The most common variants of CTCL include mycosis fungoides (MF), Sézary syndrome (SS) and CD30+ Lymphoproliferative disorders (CD30+ LPDs). CD30+ LPDs include primary cutaneous anaplastic large cell lymphoma (pcALCL), lymphomatoid papulosis (LyP) and borderline CD30+ LPD. The frequency of MF, SS and CD30+ LPDs is ~40–50%, <5% and ~10–25%, respectively. Despite recent advances, CTCL remains challenging to diagnose. The mechanism of CTCL carcinogenesis still remains to be fully elucidated. Hence, experiments in patient-derived cell lines and xenografts/genetically engineered mouse models (GEMMs) are critical to advance our understanding of disease pathogenesis. To enable this, understanding the intricacies and limitations of each individual model system is highly important. Presently, 11 immortalized patient-derived cell lines and different xenograft/GEMMs are being used to study the pathogenesis of CTCL and evaluate the therapeutic efficacy of various treatment modalities prior to clinical trials. Gene expression studies, and the karyotyping analyses of cell lines demonstrated that the molecular profile of SeAx, Sez4, SZ4, H9 and Hut78 is consistent with SS origin; MyLa and HH resemble the molecular profile of advanced MF, while Mac2A and PB2B represent CD30+ LPDs. Molecular analysis of the other two frequently used Human T-Cell Lymphotropic Virus-1 (HTLV-1)+ cell lines, MJ and Hut102, were found to have characteristics of Adult T-cell Leukemia/Lymphoma (ATLL). Studies in mouse models demonstrated that xenograft tumors could be grown using MyLa, HH, H9, Hut78, PB2B and SZ4 cells in NSG (NOD Scid gamma mouse) mice, while several additional experimental GEMMs were established to study the pathogenesis, effect of drugs and inflammatory cytokines in CTCL. The current review summarizes cell lines and xenograft/GEMMs used to study and understand the etiology and heterogeneity of CTCL.
Collapse
Affiliation(s)
- Raman Preet Kaur Gill
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Jennifer Gantchev
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Amelia Martínez Villarreal
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Brandon Ramchatesingh
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Elena Netchiporouk
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Oleg E. Akilov
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Niels Ødum
- Division of Dermatology, University of Alberta, Edmonton, AB T6G 2B7, Canada;
| | - Robert Gniadecki
- Skin Immunology Research Center, University of Copenhagen, DK-2200 Copenhagen, Denmark;
| | - Sergei B. Koralov
- Department of Pathology, New York University, New York, NY 10016, USA;
| | - Ivan V. Litvinov
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
- Correspondence: ; Tel.: +514-934-1934 (ext. 76140); Fax: +514-843-1570
| |
Collapse
|
18
|
Gökşin Ş, İmren IG, Cenk H, Kaçar N, Duygulu Ş. The Use of Interferon‐α2a as Monotherapy in Stage
IB
Patients with Mycosis Fungoides: A Retrospective Chart Review of Patient Outcomes. Dermatol Ther 2022; 35:e15344. [DOI: 10.1111/dth.15344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/28/2021] [Accepted: 01/27/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Şule Gökşin
- Pamukkale University Medical Faculty, Dermatology Department Denizli
| | - Işıl Göğem İmren
- Pamukkale University Medical Faculty, Dermatology Department Denizli
| | - Hülya Cenk
- Pamukkale University Medical Faculty, Dermatology Department Denizli
| | - Nida Kaçar
- Pamukkale University Medical Faculty, Dermatology Department Denizli
| | - Şeniz Duygulu
- Pamukkale University Medical Faculty, Dermatology Department Denizli
| |
Collapse
|
19
|
Liu Z, Wu X, Hwang ST, Liu J. The Role of Tumor Microenvironment in Mycosis Fungoides and Sézary Syndrome. Ann Dermatol 2021; 33:487-496. [PMID: 34858000 PMCID: PMC8577908 DOI: 10.5021/ad.2021.33.6.487] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 11/25/2022] Open
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are the most common subtypes of cutaneous T-cell lymphomas (CTCLs). Most cases of MF display an indolent course during its early stage. However, in some patients, it can progress to the tumor stage with potential systematic involvement and a poor prognosis. SS is defined as an erythrodermic CTCL with leukemic involvements. The pathogenesis of MF and SS is still not fully understood, but recent data have found that the development of MF and SS is related to genetic alterations and possibly to environmental influences. In CTCL, many components interacting with tumor cells, such as tumor-associated macrophages, fibroblasts, dendritic cells, mast cells, and myeloid-derived suppressor cells, as well as with chemokines, cytokines and other key players, establish the tumor microenvironment (TME). In turn, the TME regulates tumor cell migration and proliferation directly and indirectly and may play a critical role in the progression of MF and SS. The TME of MF and SS appear to show features of a Th2 phenotype, thus dampening tumor-related immune responses. Recently, several studies have been published on the immunological characteristics of MF and SS, but a full understanding of the CTCL-related TME remains to be determined. This review focuses on the role of the TME in MF and SS, aiming to further demonstrate the pathogenesis of the disease and to provide new ideas for potential treatments targeted at the microenvironment components of the tumor.
Collapse
Affiliation(s)
- Zhaorui Liu
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Xuesong Wu
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Sam T Hwang
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Jie Liu
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
20
|
Brumfiel CM, Patel MH, Puri P, Besch-Stokes J, Lester S, Rule WG, Khera N, Sluzevich JC, DiCaudo DJ, Comfere N, Bennani NN, Rosenthal AC, Pittelkow MR, Mangold AR. How to Sequence Therapies in Mycosis Fungoides. Curr Treat Options Oncol 2021; 22:101. [PMID: 34570278 DOI: 10.1007/s11864-021-00899-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2021] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT Choice of therapy in mycosis fungoides is based on both patient- and lymphoma-specific factors, such as disease characteristics, comorbidities, symptoms and effect on quality of life, potential associated toxicities of therapy, response and tolerance to prior lines of therapy, and convenience and practicality. Generally, we sequence therapies from least toxic, targeted, nonimmunosuppressive to more toxic, immunosuppressive and from single agent to multiple agents, as necessary. If more toxic, immunosuppressive agents are required to alleviate disease burden or symptoms, we generally use them just long enough to control the disease, then transition to a maintenance regimen with less toxic, less immunosuppressive agents.
Collapse
Affiliation(s)
- Caitlin M Brumfiel
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Meera H Patel
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Pranav Puri
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Jake Besch-Stokes
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Scott Lester
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - William G Rule
- Department of Radiation Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Nandita Khera
- Division of Hematology Oncology, Mayo Clinic, Phoenix, AZ, USA
| | | | - David J DiCaudo
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Nneka Comfere
- Department of Dermatology, Mayo Clinic, Rochester, MN, USA
| | - N Nora Bennani
- Division of Hematology Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Mark R Pittelkow
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Aaron R Mangold
- Department of Dermatology, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ, 85259, USA.
| |
Collapse
|
21
|
Horwitz S, Zinzani PL, Bagot M, Kim YH, Moskowitz AJ, Porcu P, Dwyer K, Sun W, Herr FM, Scarisbrick J. Lack of impact of type and extent of prior therapy on outcomes of mogamulizumab therapy in patients with cutaneous T cell lymphoma in the MAVORIC trial. Leuk Lymphoma 2021; 62:3109-3118. [PMID: 34304674 PMCID: PMC9447791 DOI: 10.1080/10428194.2021.1953007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Patients with mycosis fungoides (MF) and Sézary syndrome (SS) often require multiple lines of systemic therapy. In the phase 3 MAVORIC study (NCT01728805), mogamulizumab demonstrated superiority to vorinostat in median progression-free survival (PFS) and confirmed overall response rate (ORR) in patients with MF/SS. This post hoc analysis examined the effects of number and type of prior systemic therapies on mogamulizumab response. MAVORIC patients randomized to mogamulizumab (1.0 mg/kg intravenously weekly) or vorinostat (400 mg orally daily) were grouped by number of prior therapies and immunomodulatory activity of immediate prior systemic therapy while also considering time elapsed since treatment. ORR, PFS, and duration of response (DOR) did not vary with number of prior therapies. ORR and DOR remained consistent regardless of immediate prior therapy type. Additionally, immunomodulatory activity of the last prior therapy and time from prior treatment generally did not affect the ORR or PFS observed in response to mogamulizumab.
Collapse
Affiliation(s)
- Steven Horwitz
- Lymphoma Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia 'Seràgnoli', Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, Bologna, Italia
| | - Martine Bagot
- Service de Dermatologie, Hôpital Saint Louis, Université de Paris, Paris, France
| | - Youn H Kim
- Multidisciplinary Cutaneous Lymphoma Program, Stanford Cancer Institute, Stanford, CA, USA
| | - Alison J Moskowitz
- Lymphoma Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Pierluigi Porcu
- Department of Medical Oncology, Sidney Kimmel Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Karen Dwyer
- Medical Sciences, Kyowa Kirin, Inc, Princeton, NJ, USA
| | - Wei Sun
- Biostatistics, Kyowa Kirin, Inc, Princeton, NJ, USA
| | - Fiona M Herr
- Medical Affairs, Kyowa Kirin, Inc, Bedminster, NJ, USA
| | | |
Collapse
|
22
|
Sanches JA, Cury-Martins J, Abreu RM, Miyashiro D, Pereira J. Mycosis fungoides and Sézary syndrome: focus on the current treatment scenario. An Bras Dermatol 2021; 96:458-471. [PMID: 34053802 PMCID: PMC8245718 DOI: 10.1016/j.abd.2020.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 11/28/2022] Open
Abstract
Cutaneous T-cell lymphomas are a heterogeneous group of lymphoproliferative disorders, characterized by infiltration of the skin by mature malignant T cells. Mycosis fungoides is the most common form of cutaneous T-cell lymphoma, accounting for more than 60% of cases. Mycosis fungoides in the early-stage is generally an indolent disease, progressing slowly from some patches or plaques to more widespread skin involvement. However, 20% to 25% of patients progress to advanced stages, with the development of skin tumors, extracutaneous spread and poor prognosis. Treatment modalities can be divided into two groups: skin-directed therapies and systemic therapies. Therapies targeting the skin include topical agents, phototherapy and radiotherapy. Systemic therapies include biological response modifiers, immunotherapies and chemotherapeutic agents. For early-stage mycosis fungoides, skin-directed therapies are preferred, to control the disease, improve symptoms and quality of life. When refractory or in advanced-stage disease, systemic treatment is necessary. In this article, the authors present a compilation of current treatment options for mycosis fungoides and Sézary syndrome.
Collapse
Affiliation(s)
- José Antonio Sanches
- Dermatology Clinic Division, Faculty of Medicine, Hospital das Clínicas, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Jade Cury-Martins
- Dermatology Clinic Division, Faculty of Medicine, Hospital das Clínicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Denis Miyashiro
- Dermatology Clinic Division, Faculty of Medicine, Hospital das Clínicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Juliana Pereira
- Hematology Clinic Division, Faculty of Medicine, Hospital das Clínicas, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
23
|
Sethi TK, Montanari F, Foss F, Reddy N. How we treat advanced stage cutaneous T-cell lymphoma - mycosis fungoides and Sézary syndrome. Br J Haematol 2021; 195:352-364. [PMID: 33987825 DOI: 10.1111/bjh.17458] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
T-cell lymphomas (TCLs) constitute a rare subset of non-Hodgkin lymphomas, with mycosis fungoides/Sézary syndrome (MF/SS) being the most common subtype of cutaneous TCLs (CTCLs). Considered an incurable but treatable disease, MF/SS management presents several challenges including diagnostic delays, debilitating effect on patients' quality of life, need for several lines of therapies, multidisciplinary care and cumulative drug toxicities limiting duration of use. The present review intends to provide an overview of the recent advances in our understanding of the biology of CTCL and how these are being leveraged to provide additional treatment options for management of advanced and recurrent disease. In addition, the discussion of the different modalities of treatment is summarised to further outline the importance of multidisciplinary care and early referral to CTCL centres.
Collapse
Affiliation(s)
- Tarsheen K Sethi
- Division of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Francesca Montanari
- Division of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Francine Foss
- Division of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Nishitha Reddy
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
24
|
Mehdi SJ, Moerman-Herzog A, Wong HK. Normal and cancer fibroblasts differentially regulate TWIST1, TOX and cytokine gene expression in cutaneous T-cell lymphoma. BMC Cancer 2021; 21:492. [PMID: 33941102 PMCID: PMC8091512 DOI: 10.1186/s12885-021-08142-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma (CTCL) that transforms from mature, skin-homing T cells and progresses during the early stages in the skin. The role of the skin microenvironment in MF development is unclear, but recent findings in a variety of cancers have highlighted the role of stromal fibroblasts in promoting or inhibiting tumorigenesis. Stromal fibroblasts are an important part of the cutaneous tumor microenvironment (TME) in MF. Here we describe studies into the interaction of TME-fibroblasts and malignant T cells to gain insight into their role in CTCL. METHODS Skin from normal (n = 3) and MF patients (n = 3) were analyzed for FAPα by immunohistochemistry. MyLa is a CTCL cell line that retains expression of biomarkers TWIST1 and TOX that are frequently detected in CTCL patients. MyLa cells were cultured in the presence or absence of normal or MF skin derived fibroblasts for 5 days, trypsinized to detached MyL a cells, and gene expression analyzed by RT-PCR for MF biomarkers (TWIST1 and TOX), Th1 markers (IFNG, TBX21), Th2 markers (GATA3, IL16), and proliferation marker (MKI67). Purified fibroblasts were assayed for VIM and ACTA2 gene expression. Cellular senescence assay was performed to assess senescence. RESULTS MF skin fibroblast showed increased expression of FAP-α with increasing stage compared to normal. Normal fibroblasts co-cultured with MyLa cells suppressed expression of TWIST1 (p < 0.0006), and TOX (p < 0.03), GATA3 (p < 0.02) and IL16 (p < 0.03), and increased expression of IFNG (p < 0.03) and TBX21 (p < 0.03) in MyLa cells. In contrast, MyLa cells cultured with MF fibroblasts retained high expression of TWIST1, TOX and GATA3. MF fibroblasts co-culture with MyLa cells increased expression of IL16 (p < 0.01) and IL4 (p < 0.02), and suppressed IFNG and TBX21 in MyLa cells. Furthermore, expression of MKI67 in MyLa cells was suppressed by normal fibroblasts compared to MF fibroblasts. CONCLUSION Skin fibroblasts represent important components of the TME in MF. In co-culture model, normal and MF fibroblasts have differential influence on T-cell phenotype in modulating expression of Th1 cytokine and CTCL biomarker genes to reveal distinct roles with implications in MF progression.
Collapse
Affiliation(s)
- Syed Jafar Mehdi
- Department of Dermatology, University of Arkansas for Medical Sciences, 4301 West Markham St, #576, Little Rock, AR, 72205, USA
| | - Andrea Moerman-Herzog
- Department of Dermatology, University of Arkansas for Medical Sciences, 4301 West Markham St, #576, Little Rock, AR, 72205, USA
| | - Henry K Wong
- Department of Dermatology, University of Arkansas for Medical Sciences, 4301 West Markham St, #576, Little Rock, AR, 72205, USA.
| |
Collapse
|
25
|
Topical and Systemic Formulation Options for Cutaneous T Cell Lymphomas. Pharmaceutics 2021; 13:pharmaceutics13020200. [PMID: 33540765 PMCID: PMC7913115 DOI: 10.3390/pharmaceutics13020200] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/17/2022] Open
Abstract
Although various anti-cutaneous T-cell lymphoma (CTCL) therapies are available for clinical use, appropriate chemotherapy lines for the treatment of CTCLs have yet to be established. Therefore, to date, various clinical trials for the treatment of advanced CTCLs are ongoing. In this review, we evaluate the therapeutic options that are available in clinical practice for treatment of early- and advanced-stage CTCLs (targeted therapies, histone deacetylase (HDAC) inhibitors, retinoids, interferons, cytotoxic drugs, etc.). We also examine clinical trials of novel regimens for the treatment of CTCLs.
Collapse
|
26
|
Sun D, Zheng S, Hong YX, Chen HD, Gao XH. Subcutaneous panniculitis-like T cell lymphoma presented as erythema nodosum: A case report. Dermatol Ther 2020; 34:e14572. [PMID: 33219732 DOI: 10.1111/dth.14572] [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: 08/16/2020] [Revised: 10/27/2020] [Accepted: 11/15/2020] [Indexed: 11/28/2022]
Abstract
Subcutaneous panniculitis-like T cell lymphoma (SPTCL) is an extremely rare subtype of primary cutaneous T cell lymphomas mimicking panniculitis. Clinically, patients are usually presented with subcutaneous nodules, which usually leads to initial misdiagnosis as a benign cutaneous condition. Here, we report a 40-year-old female who presented with subcutaneous erythematous nodules on her extremities with fever. On the basis of the clinical presentations, histopathological features and immunohistochemical findings, a diagnosis of SPTCL was made. The patient was treated with the injection of recombinant human interferon α-1b (30 μg) every other day for 3 months. The lesions gradually regressed. No new erythema nodules reappeared during the 10-month follow-up.
Collapse
Affiliation(s)
- Donghong Sun
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Song Zheng
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Yu-Xiao Hong
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Hong-Duo Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Xing-Hua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Photochemotherapy Induces Interferon Type III Expression via STING Pathway. Cells 2020; 9:cells9112452. [PMID: 33182724 PMCID: PMC7697763 DOI: 10.3390/cells9112452] [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: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 11/16/2022] Open
Abstract
DNA-damaging cancer therapies induce interferon expression and stimulate the immune system, promoting therapy responses. The immune-activating STING (Stimulator of Interferon Genes) pathway is induced when DNA or double-stranded RNA (dsRNA) is detected in the cell cytoplasm, which can be caused by viral infection or by DNA damage following chemo- or radiotherapy. Here, we investigated the responses of cutaneous T-cell lymphoma (CTCL) cells to the clinically applied DNA crosslinking photochemotherapy (combination of 8–methoxypsoralen and UVA light; 8–MOP + UVA). We showed that this treatment evokes interferon expression and that the type III interferon IFNL1 is the major cytokine induced. IFNL1 upregulation is dependent on STING and on the cytoplasmic DNA sensor cyclic GMP-AMP synthase (cGAS). Furthermore, 8–MOP + UVA treatment induced the expression of genes in pathways involved in response to the tumor necrosis factor, innate immune system and acute inflammatory response. Notably, a subset of these genes was under control of the STING–IFNL1 pathway. In conclusion, our data connected DNA damage with immune system activation via the STING pathway and contributed to a better understanding of the effectiveness of photochemotherapy.
Collapse
|
28
|
Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X, Cao K, Deng H, He Y, Liao Q, Xiang B, Zhou M, Guo C, Zeng Z, Li G, Li X, Xiong W. The role of microenvironment in tumor angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:204. [PMID: 32993787 PMCID: PMC7526376 DOI: 10.1186/s13046-020-01709-5] [Citation(s) in RCA: 338] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Tumor angiogenesis is necessary for the continued survival and development of tumor cells, and plays an important role in their growth, invasion, and metastasis. The tumor microenvironment—composed of tumor cells, surrounding cells, and secreted cytokines—provides a conducive environment for the growth and survival of tumors. Different components of the tumor microenvironment can regulate tumor development. In this review, we have discussed the regulatory role of the microenvironment in tumor angiogenesis. High expression of angiogenic factors and inflammatory cytokines in the tumor microenvironment, as well as hypoxia, are presumed to be the reasons for poor therapeutic efficacy of current anti-angiogenic drugs. A combination of anti-angiogenic drugs and antitumor inflammatory drugs or hypoxia inhibitors might improve the therapeutic outcome.
Collapse
Affiliation(s)
- Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Jie Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiangying Deng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Fang Xiong
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Cao
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yi He
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| |
Collapse
|
29
|
Bobrowicz M, Fassnacht C, Ignatova D, Chang YT, Dimitriou F, Guenova E. Pathogenesis and Therapy of Primary Cutaneous T-Cell Lymphoma: Collegium Internationale Allergologicum (CIA) Update 2020. Int Arch Allergy Immunol 2020; 181:733-745. [PMID: 32690848 DOI: 10.1159/000509281] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 11/19/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a heterogeneous disease group of unknown etiology with a complex immunological background. As CTCL arises from T cells that have a vital role in the antitumor response, their therapy is largely aimed at reversing the immunological mechanisms leading to or manifesting during this malignancy. Early disease stages can be controlled with skin-directed therapy in most CTCL cases. Still, advanced CTCL has a dismal prognosis and warrants systemic therapy. Despite considerable progress in understanding the pathophysiology of the disease and the numerous systemic treatment options available, long-term remission rates with conventional treatments alone are still low. Allogeneic hematopoietic stem cell transplantation is currently the only curative option for advanced CTCL, including mycosis fungoides and Sézary syndrome. The aims of this review is to summarize the recent findings on the immunology of this heterogeneous disease and to present the advances in its clinical management.
Collapse
Affiliation(s)
| | - Christina Fassnacht
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Desislava Ignatova
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Yun-Tsan Chang
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Department of Dermatology, Lausanne University Hospital (CHUV), Lausanne, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland, .,Faculty of Medicine, University of Zurich, Zurich, Switzerland, .,Department of Dermatology, Lausanne University Hospital (CHUV), Lausanne, Switzerland, .,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,
| |
Collapse
|
30
|
Abstract
BACKGROUND Mycosis fungoides (MF) is the most common type of cutaneous T-cell lymphoma, a malignant, chronic disease initially affecting the skin. Several therapies are available, which may induce clinical remission for a time. This is an update of a Cochrane Review first published in 2012: we wanted to assess new trials, some of which investigated new interventions. OBJECTIVES To assess the effects of interventions for MF in all stages of the disease. SEARCH METHODS We updated our searches of the following databases to May 2019: the Cochrane Skin Specialised Register, CENTRAL, MEDLINE, Embase, and LILACS. We searched 2 trials registries for additional references. For adverse event outcomes, we undertook separate searches in MEDLINE in April, July and November 2017. SELECTION CRITERIA Randomised controlled trials (RCTs) of local or systemic interventions for MF in adults with any stage of the disease compared with either another local or systemic intervention or with placebo. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. The primary outcomes were improvement in health-related quality of life as defined by participants, and common adverse effects of the treatments. Key secondary outcomes were complete response (CR), defined as complete disappearance of all clinical evidence of disease, and objective response rate (ORR), defined as proportion of patients with a partial or complete response. We used GRADE to assess the certainty of evidence and considered comparisons of psoralen plus ultraviolet A (PUVA) light treatment as most important because this is first-line treatment for MF in most guidelines. MAIN RESULTS This review includes 20 RCTs (1369 participants) covering a wide range of interventions. The following were assessed as either treatments or comparators: imiquimod, peldesine, hypericin, mechlorethamine, nitrogen mustard and intralesional injections of interferon-α (IFN-α) (topical applications); PUVA, extracorporeal photopheresis (ECP: photochemotherapy), and visible light (light applications); acitretin, bexarotene, lenalidomide, methotrexate and vorinostat (oral agents); brentuximab vedotin; denileukin diftitox; mogamulizumab; chemotherapy with cyclophosphamide, doxorubicin, etoposide, and vincristine; a combination of chemotherapy with electron beam radiation; subcutaneous injection of IFN-α; and intramuscular injections of active transfer factor (parenteral systemics). Thirteen trials used an active comparator, five were placebo-controlled, and two compared an active operator to observation only. In 14 trials, participants had MF in clinical stages IA to IIB. All participants were treated in secondary and tertiary care settings, mainly in Europe, North America or Australia. Trials recruited both men and women, with more male participants overall. Trial duration varied from four weeks to 12 months, with one longer-term study lasting more than six years. We judged 16 trials as at high risk of bias in at least one domain, most commonly performance bias (blinding of participants and investigators), attrition bias and reporting bias. None of our key comparisons measured quality of life, and the two studies that did presented no usable data. Eighteen studies reported common adverse effects of the treatments. Adverse effects ranged from mild symptoms to lethal complications depending upon the treatment type. More aggressive treatments like systemic chemotherapy generally resulted in more severe adverse effects. In the included studies, CR rates ranged from 0% to 83% (median 31%), and ORR ranged from 0% to 88% (median 47%). Five trials assessed PUVA treatment, alone or combined, summarised below. There may be little to no difference between intralesional IFN-α and PUVA compared with PUVA alone for 24 to 52 weeks in CR (risk ratio (RR) 1.07, 95% confidence interval (CI) 0.87 to 1.31; 2 trials; 122 participants; low-certainty evidence). Common adverse events and ORR were not measured. One small cross-over trial found once-monthly ECP for six months may be less effective than twice-weekly PUVA for three months, reporting CR in two of eight participants and ORR in six of eight participants after PUVA, compared with no CR or ORR after ECP (very low-certainty evidence). Some participants reported mild nausea after PUVA but no numerical data were given. One participant in the ECP group withdrew due to hypotension. However, we are unsure of the results due to very low-certainty evidence. One trial comparing bexarotene plus PUVA versus PUVA alone for up to 16 weeks reported one case of photosensitivity in the bexarotene plus PUVA group compared to none in the PUVA-alone group (87 participants; low-certainty evidence). There may be little to no difference between bexarotene plus PUVA and PUVA alone in CR (RR 1.41, 95% CI 0.71 to 2.80) and ORR (RR 0.94, 95% CI 0.61 to 1.44) (93 participants; low-certainty evidence). One trial comparing subcutaneous IFN-α injections combined with either acitretin or PUVA for up to 48 weeks or until CR indicated there may be little to no difference in the common IFN-α adverse effect of flu-like symptoms (RR 1.32, 95% CI 0.92 to 1.88; 82 participants). There may be lower CR with IFN-α and acitretin compared with IFN-α and PUVA (RR 0.54, 95% CI 0.35 to 0.84; 82 participants) (both outcomes: low-certainty evidence). This trial did not measure ORR. One trial comparing PUVA maintenance treatment to no maintenance treatment, in participants who had already had CR, did report common adverse effects. However, the distribution was not evaluable. CR and OR were not assessable. The range of treatment options meant that rare adverse effects consequently occurred in a variety of organs. AUTHORS' CONCLUSIONS There is a lack of high-certainty evidence to support decision making in the treatment of MF. Because of substantial heterogeneity in design, missing data, small sample sizes, and low methodological quality, the comparative safety and efficacy of these interventions cannot be reliably established on the basis of the included RCTs. PUVA is commonly recommended as first-line treatment for MF, and we did not find evidence to challenge this recommendation. There was an absence of evidence to support the use of intralesional IFN-α or bexarotene in people receiving PUVA and an absence of evidence to support the use of acitretin or ECP for treating MF. Future trials should compare the safety and efficacy of treatments to PUVA, as the current standard of care, and should measure quality of life and common adverse effects.
Collapse
Affiliation(s)
- Arash Valipour
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University Hospital, Frankfurt am Main, Germany
- Evidence-Based Medicine Frankfurt, Institute of General Practice, Goethe University, Frankfurt, Germany
| | - Manuel Jäger
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University Hospital, Frankfurt am Main, Germany
- Hautklinik, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Peggy Wu
- Department of Dermatology, University of California Davis, Sacramento, CA, USA
| | - Jochen Schmitt
- Center for Evidence-Based Healthcare, Faculty of Medicine Carl Gustav Carus, Technischen Universität (TU) Dresden, Dresden, Germany
| | - Charles Bunch
- c/o Cochrane Skin Group, The University of Nottingham, Nottingham, UK
| | - Tobias Weberschock
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University Hospital, Frankfurt am Main, Germany
- Evidence-Based Medicine Frankfurt, Institute of General Practice, Goethe University, Frankfurt, Germany
| |
Collapse
|
31
|
Ferranti M, Tadiotto Cicogna G, Russo I, Alaibac M. Immunomodulation in Cutaneous T Cell Lymphoma. Front Oncol 2019; 9:1025. [PMID: 31649883 PMCID: PMC6794804 DOI: 10.3389/fonc.2019.01025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 09/23/2019] [Indexed: 12/14/2022] Open
Affiliation(s)
| | | | - Irene Russo
- Unit of Dermatology, University of Padua, Padua, Italy
| | - Mauro Alaibac
- Unit of Dermatology, University of Padua, Padua, Italy
| |
Collapse
|
32
|
Sivanand A, Surmanowicz P, Alhusayen R, Hull P, Litvinov IV, Zhou Y, Gniadecki R. Immunotherapy for Cutaneous T-Cell Lymphoma: Current Landscape and Future Developments. J Cutan Med Surg 2019; 23:537-544. [PMID: 31353944 DOI: 10.1177/1203475419867610] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are chronic, progressive primary cutaneous T-cell lymphomas (CTCLs) for which there are no curative treatments. Skin-directed therapies, such as phototherapy, radiation therapy, or topical nitrogen mustard, provide only short-term remissions. Numerous attempts with different chemotherapeutic regimes failed to achieve meaningful clinical responses. Immunotherapy seems to be a promising avenue to achieve long-term disease control in CTCL. There is compelling evidence indicating that MF and SS are immunogenic lymphomas, which can be recognized by the patient's immune system. However, CTCL uses different strategies to impair host's immunity, eg, via repolarizing the T-cell differentiation from type I to type II, recruiting immunosuppressive regulatory T-cells, and limiting the repertoire of lymphocytes in the circulation. Many currently used therapies, such as interferon-α, imiquimod, extracorporeal phototherapy, and allogeneic bone marrow transplant, seem to exert their therapeutic effect via activation of the antitumor cytotoxic response and reconstitution of the host's immune system. It is likely that novel immunotherapies such as immune checkpoint inhibitors, cancer vaccines, and chimeric antigen receptor-T cells will help to manage CTCL more efficiently. We also discuss how current genomic techniques, such as estimating the mutational load by whole genome sequencing and neoantigen calling, are likely to provide clinically useful information facilitating personalized immunotherapy of CTCL.
Collapse
Affiliation(s)
- Arunima Sivanand
- 1 Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | | | - Raed Alhusayen
- 2 Division of Dermatology, University of Toronto, ON, Canada
| | - Peter Hull
- 3 Division of Clinical Dermatology and Cutaneous Science, Dalhousie University, Halifax, NS, Canada
| | - Ivan V Litvinov
- 4 Division of Dermatology, McGill University, Montreal, QC, Canada
| | - Youwen Zhou
- 5 Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
| | - Robert Gniadecki
- 1 Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
33
|
Doerschner M, Pekar-Lukacs A, Messerli-Odermatt O, Dommann-Scherrer C, Rütti M, Müller AM, Nair G, Kamarachev J, Kerl K, Beer M, Messerli M, Frauenknecht K, Haralambieva E, Hoetzenecker W, French LE, Guenova E. Interferon alfa-2a maintenance after salvage autologous stem cell transplantation in atypical mycosis fungoides with central nervous system involvement. Br J Dermatol 2019; 181:1296-1302. [PMID: 30565216 DOI: 10.1111/bjd.17535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2018] [Indexed: 01/05/2023]
Abstract
Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma with unfavourable prognosis for patients with advanced stages of the disease. Refractory disease and advanced-stage disease require systemic therapy. We report on a rare case of an atypical predominantly CD8+ folliculotropic MF, a subtype of MF with poorer prognosis, in a 59-year-old woman. She was initially diagnosed with MF restricted to the skin, of T3N0M0B0/stage IIB according to the current World Health Organization-European Organisation for Research and Treatment of Cancer classification. First-line treatment with local percutaneous radiotherapy in combination with systemic interferon alfa-2a resulted in complete remission. However, 21 months later the disease progressed to T3N0M1B0/stage IVB with development of cerebral manifestation and thus very poor prognosis. Allogeneic stem cell transplantation (SCT) was not a therapeutic option due to the lack of a suitable donor. We initiated methotrexate and cytarabine chemotherapy, followed by high-dose chemotherapy with thiotepa and carmustine with autologous SCT. Despite rapid response and complete remission of the cerebral lesions, disease recurrence of the skin occurred soon after. Interestingly, readministration of interferon alfa-2a as a maintenance treatment after the salvage autologous SCT resulted in a durable complete remission during the follow-up period of currently 17 months after autologous SCT. What's already known about this topic? Mycosis fungoides is a primary cutaneous T-cell lymphoma with unfavourable prognosis for the advanced stages of the disease. A refractory course of disease requires systemic therapy. What does this study add? We report on an unusual case of a patient with mycosis fungoides with cerebral involvement, in which a durable complete remission was achieved upon autologous stem cell therapy and interferon alfa-2a maintenance therapy.
Collapse
Affiliation(s)
- M Doerschner
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - A Pekar-Lukacs
- Department of Oncology and Pathology, Lund University, Lund, Sweden
| | | | - C Dommann-Scherrer
- Institute of Pathology, Canton Hospital Winterthur, Winterthur, Switzerland
| | - M Rütti
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - A M Müller
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - G Nair
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - J Kamarachev
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - K Kerl
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - M Beer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - M Messerli
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - K Frauenknecht
- Institute of Neuropathology, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - E Haralambieva
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - W Hoetzenecker
- Department of Dermatology, University Hospital Linz, Linz, Austria
| | - L E French
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - E Guenova
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, Wu X, Ma J, Zhou M, Li X, Li Y, Li G, Xiong W, Guo C, Zeng Z. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer 2019; 18:10. [PMID: 30646912 PMCID: PMC6332843 DOI: 10.1186/s12943-018-0928-4] [Citation(s) in RCA: 854] [Impact Index Per Article: 170.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/26/2018] [Indexed: 12/14/2022] Open
Abstract
Tumor immune escape is an important strategy of tumor survival. There are many mechanisms of tumor immune escape, including immunosuppression, which has become a research hotspot in recent years. The programmed death ligand-1/programmed death-1 (PD-L1/PD-1) signaling pathway is an important component of tumor immunosuppression, which can inhibit the activation of T lymphocytes and enhance the immune tolerance of tumor cells, thereby achieving tumor immune escape. Therefore, targeting the PD-L1/PD-1 pathway is an attractive strategy for cancer treatment; however, the therapeutic effectiveness of PD-L1/PD-1 remains poor. This situation requires gaining a deeper understanding of the complex and varied molecular mechanisms and factors driving the expression and activation of the PD-L1/PD-1 signaling pathway. In this review, we summarize the regulation mechanisms of the PD-L1/PD-1 signaling pathway in the tumor microenvironment and their roles in mediating tumor escape. Overall, the evidence accumulated to date suggests that induction of PD-L1 by inflammatory factors in the tumor microenvironment may be one of the most important factors affecting the therapeutic efficiency of PD-L1/PD-1 blocking.
Collapse
Affiliation(s)
- Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Jie Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Xiangying Deng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Fang Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xu Wu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Department of Chemistry, University of North Dakota, Grand Forks, North Dakota, 58202, USA
| | - Jian Ma
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Yong Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, 410078, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
35
|
Wain T, Pavli A, Wells J, Fernandez-Peñas P. The efficacy and safety of methotrexate versus interferon in cutaneous T-cell lymphomas. J DERMATOL TREAT 2018; 29:715-719. [PMID: 29455635 DOI: 10.1080/09546634.2018.1441492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Methotrexate (MTX) and interferon (IFN) have been used in the treatment of cutaneous T-cell lymphomas (CTCL) of various subtypes. We review our experience of MTX and IFN use in our patients with CTCL at a tertiary hospital. MATERIALS AND METHODS Medical records of patients over 4 years were reviewed. We describe the dosages, time to response, response rates, side effects, progression rate, and reasons for discontinuation. RESULTS Response rate was significantly higher in the IFN group than MTX group (86.67% and 47.4% respectively, p = .01). Disease progression occurred 57.89% in the MTX group whilst only 26.67% progressed with IFN therapy. Patients taking IFN therapy experienced proportionally more side effects of any type than those undertaking MTX treatment (86.67% vs. 47.37%, odds ratio 7.22). However, discontinuation rate in the IFN group (26.67%) was much lower than in the MTX arm (89.47%). CONCLUSIONS The most significant finding of this study was that patients with CTCL treated with IFN had a better response rate and significantly shorter response time compared with those treated with MTX. Additionally, patients had less disease progression on IFN than with MTX regardless of subtype of T-cell lymphoma and stage of disease.
Collapse
Affiliation(s)
- Thevaki Wain
- a Department of Dermatology , Westmead Hospital , Sydney , Australia
| | - Alexandra Pavli
- a Department of Dermatology , Westmead Hospital , Sydney , Australia
| | - Jillian Wells
- a Department of Dermatology , Westmead Hospital , Sydney , Australia
| | - Pablo Fernandez-Peñas
- a Department of Dermatology , Westmead Hospital , Sydney , Australia.,b Sydney Medical School , University of Sydney , Sydney , Australia
| |
Collapse
|
36
|
ManfrereC KCG, Torrealba MP, Miyashiro DR, Pereira NZ, Yoshikawa FSY, de M Oliveira L, Cury-Martins J, Duarte AJS, Sanches JA, Sato MN. Profile of differentially expressed Toll-like receptor signaling genes in the natural killer cells of patients with Sézary syndrome. Oncotarget 2017; 8:92183-92194. [PMID: 29190907 PMCID: PMC5696173 DOI: 10.18632/oncotarget.21006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/27/2017] [Indexed: 11/25/2022] Open
Abstract
Sézary syndrome (SS), an aggressive and leukemic form of cutaneous T-cell lymphoma, usually results in shortened survival. Improving innate immunity in SS by targeting natural killer (NK) cells with Toll-like receptor (TLR) agonists could be an interesting modulatory strategy. We evaluated the NK cell populations in SS patients assessing activating and inhibitory receptors expression and profiled the differential expression of TLR signaling pathway genes in unstimulated NK cells and after TLR7/8 stimulation. We observed preserved CD56bright NK cells and a low percentage of CD56dim NK cells in the peripheral blood of SS patients compared to those in the healthy control group. Both NK cell populations showed down-modulation of NKG2C and NKG2D expression, which was associated with high serum levels of the soluble form of NKG2D ligands. In contrast, an expansion of “memory” CD57+ NKG2C+ NK cells and high cytomegalovirus antibody titers were detected in SS patients. Profiling of the TLR signaling genes in NK cells from SS patients showed an abundance of differentially expressed genes (DEGs) in NK cells in the unstimulated condition, with mostly up-regulation of NFκB/JNK p38 pathway genes, but there was down-regulation of type I (IFN-α/β) and II (IFN-γ) interferon and IL-12A. After activation of NK cells with TLR7/8 agonist, the down-regulated genes correlated with the IFN response, and IL-12 became up-regulated, together with other antitumor factors. NK cell activation with a dual agonist for TLR7 and TLR8 is able to induce the expression of IFN-γ and type I IFN, which can improve immunity in SS patients.
Collapse
Affiliation(s)
- Kelly C G ManfrereC
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Marina P Torrealba
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Denis R Miyashiro
- Cutaneous Lymphoma Clinic, Hospital das Clínicas, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Nátalli Z Pereira
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Fabio S Y Yoshikawa
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Luana de M Oliveira
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Jade Cury-Martins
- Cutaneous Lymphoma Clinic, Hospital das Clínicas, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Alberto J S Duarte
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - José A Sanches
- Cutaneous Lymphoma Clinic, Hospital das Clínicas, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Maria N Sato
- Laboratory of Medical Investigation, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
37
|
Stranzenbach R, Dippel E, Schlaak M, Stadler R. Alternate dosing regimens of brentuximab vedotin for CD30+ cutaneous T-cell lymphoma: reply from the authors. Br J Dermatol 2017; 178:303. [PMID: 28889619 DOI: 10.1111/bjd.15972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- R Stranzenbach
- Department of Dermatology, Venerology, Allergology and Phlebology, Johannes Wesling Medical Centre, University Hospital of Ruhr-University Bochum, Bochum, Germany
| | - E Dippel
- Department of Dermatology, Klinikum Ludwigshafen, Skin Cancer Centre Rheinpfalz, Ludwigshafen, Germany
| | - M Schlaak
- Department of Dermatology and Venereology, University Hospital Cologne, Skin Cancer Center at Center of Integrated Oncology (CIO Koeln Bonn), Cologne, Germany
| | - R Stadler
- Department of Dermatology, Venerology, Allergology and Phlebology, Johannes Wesling Medical Centre, University Hospital of Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
38
|
Lewis DJ, Kim YH, Duvic M. Alternate dosing regimens of brentuximab vedotin for CD30+ cutaneous T-cell lymphoma. Br J Dermatol 2017; 178:302-303. [PMID: 28889405 DOI: 10.1111/bjd.15970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- D J Lewis
- School of Medicine, Baylor College of Medicine, Houston, TX, U.S.A.,Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, U.S.A
| | - Y H Kim
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - M Duvic
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, U.S.A
| |
Collapse
|
39
|
Kremer KN, Dinkel BA, Sterner RM, Osborne DG, Jevremovic D, Hedin KE. TCR-CXCR4 signaling stabilizes cytokine mRNA transcripts via a PREX1-Rac1 pathway: implications for CTCL. Blood 2017; 130:982-994. [PMID: 28694325 PMCID: PMC5570680 DOI: 10.1182/blood-2017-03-770982] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/05/2017] [Indexed: 02/06/2023] Open
Abstract
As with many immunopathologically driven diseases, the malignant T cells of cutaneous T-cell lymphomas (CTCLs), such as Sézary syndrome, display aberrant cytokine secretion patterns that contribute to pathology and disease progression. Targeting this disordered release of cytokines is complicated by the changing cytokine milieu that drives the phenotypic changes of CTCLs. Here, we characterize a novel signaling pathway that can be targeted to inhibit the secretion of cytokines by modulating either CXCR4 or CXCR4-mediated signaling. We demonstrate that upon ligation of the T-cell antigen receptor (TCR), the TCR associates with and transactivates CXCR4 via phosphorylation of S339-CXCR4 in order to activate a PREX1-Rac1-signaling pathway that stabilizes interleukin-2(IL-2), IL-4, and IL-10 messenger RNA (mRNA) transcripts. Pharmacologic inhibition of either TCR-CXCR4 complex formation or PREX1-Rac1 signaling in primary human T cells decreased mRNA stability and inhibited secretion of IL-2, IL-4, and IL-10. Applying this knowledge to Sézary syndrome, we demonstrate that targeting various aspects of this signaling pathway blocks both TCR-dependent and TCR-independent cytokine secretion from a Sézary syndrome-derived cell line and patient isolates. Together, these results identify multiple aspects of a novel TCR-CXCR4-signaling pathway that could be targeted to inhibit the aberrant cytokine secretion that drives the immunopathogenesis of Sézary syndrome and other immunopathological diseases.
Collapse
MESH Headings
- Benzylamines
- Cyclams
- Cytokines/genetics
- Cytokines/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Guanine Nucleotide Exchange Factors/metabolism
- Heterocyclic Compounds/pharmacology
- Humans
- Jurkat Cells
- Lymphocyte Subsets/drug effects
- Lymphocyte Subsets/metabolism
- Lymphoma, T-Cell, Cutaneous/metabolism
- Lymphoma, T-Cell, Cutaneous/pathology
- Models, Biological
- RNA Stability/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, CXCR4/metabolism
- Sezary Syndrome/pathology
- Signal Transduction/drug effects
- Transcriptional Activation/drug effects
- Transcriptional Activation/genetics
- rac1 GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
| | | | - Rosalie M Sterner
- Department of Immunology
- Mayo Clinic Medical Scientist Training Program, and
| | | | - Dragan Jevremovic
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
40
|
Schiller M, Tsianakas A, Sterry W, Dummer R, Hinke A, Nashan D, Stadler R. Dose-escalation study evaluating pegylated interferon alpha-2a in patients with cutaneous T-cell lymphoma. J Eur Acad Dermatol Venereol 2017; 31:1841-1847. [PMID: 28557110 DOI: 10.1111/jdv.14366] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 04/28/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND This open-label, multicenter, dose-escalation study evaluated the safety, tolerability, and efficacy of subcutaneous pegylated (40 kD) interferon α-2a (PEG-IFN α-2a) in patients with cutaneous T-cell lymphoma (CTCL). PATIENTS AND METHODS PEG-IFN α-2a was administered subcutaneously at 180 (n = 4), 270 (n = 6), or 360 μg (n = 3) once weekly for 12 weeks. Efficacy was assessed by the proportion of patients with complete response (CR) or partial response (PR). RESULTS PEG-IFN α-2a was generally well tolerated, with a moderate number of reductions or withholding of doses because of adverse events (AEs) (25% (n = 1), 66% (n = 4), and 0% (n = 0) in the 180-, 270-, and 360-μg/week groups, respectively). The only dose-limiting toxicity was a grade 3 elevation of liver enzymes in the 270-μg dose group. The most common AEs were fatigue, acute flu-like symptoms, and hepatic toxicity. The major response rate (CR or PR) was 50% in the 180-μg group (CR, 50%; PR, 0%), 83% in the 270-μg group (CR, 67%; PR, 17%), and 66% in the 360-μg group (CR, 33%; PR, 33%). CONCLUSION PEG-IFN α-2a at doses up to 360 μg once weekly was well tolerated in patients with CTCL up to the highest dose group and showed good response rates. Due to their good tolerance even in high doses, they might be an option for patients not tolerating standard IFN-α preparations. However, for this purpose and to evaluate comparability between standard and PEG-IFN larger clinical trials are needed, alone and in combination with oral photochemotherapy (PUVA).
Collapse
Affiliation(s)
- M Schiller
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany.,Dermatological Office Professor Schiller, Coesfeld, Germany
| | - A Tsianakas
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - W Sterry
- Department of Dermatology, Charité, Berlin, Germany
| | - R Dummer
- Department of Dermatology, University Hospital, Zurich, Switzerland
| | - A Hinke
- WiSP Wissenschaftlicher Service Pharma GmbH, Langenfeld, Germany
| | - D Nashan
- Department of Dermatology, Klinikum Dortmund, Dortmund, Germany
| | - R Stadler
- Department of Dermatology, Johannes Wesling Klinikum Minden, University Hospital of Ruhr University of Bochum, Bochum, Germany
| |
Collapse
|
41
|
Abstract
Cutaneous T-cell lymphomas (CTCLs) are a heterogeneous group of extranodal lymphomas involving the skin. Diagnosis of the two main subtypes of CTCL-mycosis fungoides (MF) and Sézary syndrome (SS)-is based on the International Society for Cutaneous Lymphomas/European Organization for Research and Treatment of Cancer (ISCL/EORTC) classification system, which utilizes clinical, histopathological, molecular biologic, and immunopathologic features. Risk stratification, based on TNMB (tumor, node, metastasis, and blood) staging, provides prognostic information, with limited-stage disease conferring the longest median overall survival. Skin-directed therapies are preferred in the management of limited-stage disease, whereas advanced-stage disease requires systemic therapies. As the mechanisms of CTCL pathogenesis are increasingly understood, new monoclonal antibodies, checkpoint inhibitors, immunomodulatory agents, and small molecules are under investigation and may provide additional therapeutic options for those with advanced CTCL. This review examines the current landscape of targeted therapies in the treatment of CTCLs.
Collapse
Affiliation(s)
- Sumana Devata
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, 3A17 N. Ingalis Bldg, 300 N. Ingalis St. SPC 5419, Ann Arbor, MI, 48109-5419, USA.
| | - Ryan A Wilcox
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, 4310 CC, Ann Arbor, MI, 48109-5936, USA
| |
Collapse
|