251
|
Teo AYT, Yau CE, Low CE, Pereira JVB, Ng JYX, Soong TK, Lo JYT, Yang VS. Effectiveness of immune checkpoint inhibitors and other treatment modalities in patients with advanced mucosal melanomas: a systematic review and individual patient data meta-analysis. EClinicalMedicine 2024; 77:102870. [PMID: 39416390 PMCID: PMC11474374 DOI: 10.1016/j.eclinm.2024.102870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Background Mucosal melanomas (MM) are an aggressive subtype of melanoma. Given the rarity of this disease, the conduct of clinical trials is challenging and has been limited. Current treatment options have been extrapolated from the more common cutaneous melanoma even though MM is distinct in pathogenesis, etiology and prognosis. This is the first meta-analysis to comprehensively assess the efficacy of immune checkpoint inhibitors (anti-PD1 and anti-CTLA4) and other treatment modalities (targeted therapy such as KIT inhibitors and VEGF inhibitors, as well as radiotherapy) on survival outcomes in MM to develop clinical guidelines for evidence-based management. Methods The protocol was prospectively registered on PROSPERO (PROSPERO ID: CRD42023411195). PubMed, Embase, Cochrane Central Register of Controlled Trials (CENTRAL), Web of Science and Google Scholar were searched from inception until 25 July 2024, for all cohort and observational studies. Eligible studies included those with five or more participants with locally advanced or metastatic MM treated with anti-PD1, anti-CTLA4, VEGF inhibitors and/or KIT inhibitors. Titles and abstracts of potential articles were screened and full texts of all potentially eligible studies were retrieved and reviewed by two independent reviewers. Individual patient data (IPD) from published Kaplan-Meier curves were reconstructed using a graphical reconstruction method and pooled as a one-stage meta-analysis. A sensitivity analysis using a two-stage meta-analysis approach was conducted. Extracted outcomes included overall survival (OS) and progression-free survival (PFS). For each treatment arm, median survival time and 12-month survival proportion were estimated. Data from double-arm trials was pooled to estimate hazard ratios (HRs), ratios of restricted mean time lost (RMTL) and restricted mean survival time (RMST). Findings From a total of 7402 studies, 35 eligible studies comprising a total of 2833 participants were included. Combined anti-PD1 and anti-CTLA4 therapy had the highest 12-month OS and 12-month PFS at 71.8% (95% CI: 67.6%, 76.2%, n = 476) and 35.1% (95% CI: 30.5%, 40.4%, n = 401) respectively, followed by anti-PD1 therapy alone (OS: 64.0% (95% CI: 61.4%, 66.7%, n = 1399); PFS: was 28.3% (95% CI: 25.8%, 31.2%, n = 1142), anti-PD1 and VEGF inhibitor combination therapy (OS: 57.1% (95% CI: 51.0%, 63.9%)), KIT inhibitors (OS: 48.2% (95% CI: 37.6%, 61.8%); PFS: 8.3% (95% CI: 3.7%, 18.7%)) and anti-CTLA4 therapy alone (OS: 33.3% (95% CI: 28.4%, 39.1%); PFS: 9.8% (95% CI: 5.9%, 16.5%)). In the double-arm studies, combination therapy with anti-PD1 and anti-CTLA4 had similar OS and PFS with anti-PD1 alone (OS: HR 0.856 (95% CI: 0.704, 1.04); RMTL ratio 0.932 (95% CI: 0.832, 1.044, P = 0.225); RMST ratio 1.102 (95% CI: 0.948, 1.281, P = 0.204); PFS: HR 0.919 (95% CI: 0.788, 1.07); RMTL ratio 0.936 (95% CI: 0.866, 1.013, P = 0.100); RMST ratio 1.21 (95% CI: 0.979, 1.496, P = 0.078)), however, anti-PD1 therapy alone had significantly better PFS than anti-CTLA4 alone (HR 0.548 (95% CI: 0.376, 0.799); RMTL ratio 0.715 (95% CI: 0.606, 0.844, P < 0.001); RMST ratio 1.659 (95% CI: 1.316, 2.092, P < 0.001)). Anti-PD1 therapy with radiotherapy versus anti-PD1 alone showed no significant difference (OS: HR 0.854 (95% CI: 0.567, 1.29); RMTL ratio 0.855 (95% CI: 0.675, 1.083, P = 0.193); RMST ratio 1.194 (95% CI: 0.928, 1.536, P = 0.168; PFS: HR 0.994 (95% CI: 0.710, 1.39); RMTL ratio 1.006 (95% CI: 0.87, 1.162, P = 0.939); RMST ratio 0.984 (95% CI: 0.658, 1.472, P = 0.939)). Interpretation For the systemic treatment of MM, anti-PD1 is the best monotherapy. While combining anti-PD1 with other treatment options such as anti-CTLA4, VEGF inhibitors or radiotherapy might achieve better outcomes, these improvements did not reach statistical significance when evaluated by HR, RMTL and RMST ratios. Funding This work was supported by the National Medical Research Council Transition Award (TA20nov-0020), SingHealth Duke-NUS Oncology Academic Clinical Programme (08/FY2020/EX/67-A143 and 08/FY2021/EX/17-A47), the Khoo Pilot Collaborative Award (Duke-NUS-KP(Coll)/2022/0020A), the National Medical Research Council Clinician Scientist-Individual Research Grant-New Investigator Grant (CNIGnov-0025), the Terry Fox Grant (I1056) and the Khoo Bridge Funding Award (Duke-NUS-KBrFA/2024/0083I).
Collapse
Affiliation(s)
- Andrea York Tiang Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Singapore General Hospital, Singapore, 169608, Singapore
| | - Chun En Yau
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Chen Ee Low
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | | | | | | | - Jack Yu Tung Lo
- Department of Neurosurgery, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Valerie Shiwen Yang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, 138673, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, 169857, Singapore
| |
Collapse
|
252
|
Burr JL, Johnson KC, Carmicheal JJ, Lin C, Ganti AK. Combination Immunotherapy With Radiotherapy in Non-Small Cell Lung Cancer: A Review of Evidence. Cancer Med 2024; 13:e70402. [PMID: 39526426 PMCID: PMC11551781 DOI: 10.1002/cam4.70402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Radiotherapy plays a fundamental role in the treatment of patients with all stages of non-small-cell lung cancer (NSCLC). The emergence of immune checkpoint inhibitors (ICIs) has transformed the standard of care in these patients. The use of ICIs is increasingly utilized in the definitive setting as an adjunct to chemoradiotherapy or surgery and remains a vital component in the treatment of metastatic disease. Despite improvements in patient survival, the use of immunotherapy as monotherapy has shown limited overall response rates with susceptibility to resistance. Radiotherapy has been identified as a viable option to enhance the response rate to ICI and improve outcomes in NSCLC. METHODS We queried the English PubMed database utilizing variably combined search items including "radiation," "chemoradiation," "immune checkpoint," "immunotherapy," "stereotactic body radiotherapy," and "non-small-cell lung". We additionally searched various acceptable alternative terms for similar keywords such as "radiotherapy" in place of "radiation." These results were subsequently curated for relevance and impact on current treatment paradigms. RESULTS In this review, we discuss preclinical and clinical studies relating to combinatorial use of immunotherapy and radiation in NSCLC. These studies are presented in the context of early-stage, operable stage III, unresectable stage III, and metastatic disease. The majority of the data illustrate promising results regarding the additive or synergistic effects of radiation and immunotherapy with a suggestion that the timing of these treatment modalities is crucial to optimizing outcomes. CONCLUSION While there is now evidence regarding the favorable interplay between radiation and immunotherapy in NSCLC, there remain multiple unanswered questions which are expected to be addressed in ongoing clinical trials.
Collapse
Affiliation(s)
- Justin L. Burr
- Department of Radiation OncologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kurtis C. Johnson
- Department of Radiation OncologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Joseph J. Carmicheal
- Department of Radiation OncologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Chi Lin
- Department of Radiation OncologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Apar Kishor Ganti
- Division of Hematology‐Oncology, Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
253
|
Rocchetti JR, Price J. Abscopal Effect in a Patient With Metastatic Melanoma Receiving Hypofractionated Radiation Therapy and Dual Immune Checkpoint Inhibition: A Case Report. Cureus 2024; 16:e74662. [PMID: 39735011 PMCID: PMC11681968 DOI: 10.7759/cureus.74662] [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: 09/03/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Concurrent with the increasing utilization of immune checkpoint inhibitors (ICIs) in melanoma, there has been renewed interest in understanding the potential interplay between radiation therapy (RT) and the immune system. One such phenomenon is the abscopal effect, where localized treatments, such as RT, not only shrink the targeted tumor but also induce shrinkage of untreated tumors elsewhere in the body. Here, we report a case of an abscopal effect in a 63-year-old patient with metastatic melanoma who was progressing on first-line dual ICI therapy but experienced a rapid and durable systemic response following the administration of hypofractionated palliative RT to a large primary melanoma skin tumor.
Collapse
Affiliation(s)
- Jenna R Rocchetti
- Radiation Oncology, Rowan-Virtua School of Osteopathic Medicine, Stratford, USA
| | - Jeremy Price
- Radiation Oncology, Fox Chase Cancer Center, Temple University Hospital, Philadelphia, USA
| |
Collapse
|
254
|
Martineau R, Susini S, Marabelle A. Fc Effector Function of Immune Checkpoint Blocking Antibodies in Oncology. Immunol Rev 2024; 328:334-349. [PMID: 39663733 PMCID: PMC11659940 DOI: 10.1111/imr.13427] [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: 09/16/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Antagonistic monoclonal antibodies (mAbs) targeting inhibitory immune checkpoints have revolutionized the field of oncology. CTLA-4, PD-1, and LAG3 are three co-inhibitory receptors, which can be expressed by subsets of T cells and which play a role in the regulation of adaptive immune responses. Blocking these immune checkpoints receptors (or their ligands) with antagonistic antibodies can lead to tumor regressions and lasting remissions in some patients with cancer. Two anti-CTLA4, six anti-PD1, three anti-PD-L1, and one anti-LAG3 antibodies are currently approved by the FDA and EMA. Their mechanism of action, safety, and efficacy are linked to their affinity with Fc gamma receptors (FcγR) (so called "effector functions"). The anti-CTLA-4 antibodies ipilimumab (IgG1) and tremilimumab (IgG2a), and the anti-PD-L1 avelumab (IgG1) have isotypes with high affinity for activating FcγR and thereby can induce ADCC/ADCP. The effector function is required for the in vivo efficacy of anti-CTLA4 antibodies. For anti-PD(L)1 antibodies, where a pure antagonistic function ("checkpoint blockade") is sufficient, some mAbs are IgG1 but have been mutated in their Fc sequence (e.g., durvalumab and atezolizumab) or are IgG4 (e.g., nivolumab and pembrolizumab) to have low affinity for FcγR. Here, we review the impact of FcγR effector function on immune checkpoint blockers safety and efficacy in oncology.
Collapse
Affiliation(s)
- Romane Martineau
- Université Paris SaclayLe Kremlin‐BicetreFrance
- Centre d'Investigation Clinique BIOTHERIS, CIC 1428Institut National de la Santé et de la Recherche Médicale (INSERM)VillejuifFrance
| | - Sandrine Susini
- Centre d'Investigation Clinique BIOTHERIS, CIC 1428Institut National de la Santé et de la Recherche Médicale (INSERM)VillejuifFrance
- Translational Immunotherapy Research LaboratoryGustave RoussyVillejuifFrance
| | - Aurelien Marabelle
- Université Paris SaclayLe Kremlin‐BicetreFrance
- Centre d'Investigation Clinique BIOTHERIS, CIC 1428Institut National de la Santé et de la Recherche Médicale (INSERM)VillejuifFrance
- Translational Immunotherapy Research LaboratoryGustave RoussyVillejuifFrance
| |
Collapse
|
255
|
Rousseau A, Géraud A, Geiss R, Farcet A, Spano JP, Hamy AS, Gougis P. Safety of solid oncology drugs in older patients: a narrative review. ESMO Open 2024; 9:103965. [PMID: 39481329 PMCID: PMC11567126 DOI: 10.1016/j.esmoop.2024.103965] [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: 07/09/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
The older population represents ∼50%-60% of the population of newly diagnosed patients with cancer. Due to physiological and pathological aging and the increased presence of comorbidities and frailty factors, this population is at higher risk of serious toxicity from anticancer drugs and, consequently, often under-treated. Despite the complexity of these treatments, a good knowledge of the pharmacology of anticancer drugs and potentially risky situations can limit the emergence of potentially lethal toxicities in this population. This review focuses on optimizing systemic oncology treatments for older patients, emphasizing the unique characteristics of each therapeutic class and the necessity for a precautionary approach for this vulnerable population.
Collapse
Affiliation(s)
- A Rousseau
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - A Géraud
- Department of Medical Oncology, Institut Paoli-Calmette, Marseille, France
| | - R Geiss
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France
| | - A Farcet
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - J-P Spano
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - A-S Hamy
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France; Residual Tumor and Response to Treatment, RT2Lab, INSERM, U932 Cancer & Immunity, Institut Curie, Université Paris Sciences Lettres, Paris, France
| | - P Gougis
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France; Residual Tumor and Response to Treatment, RT2Lab, INSERM, U932 Cancer & Immunity, Institut Curie, Université Paris Sciences Lettres, Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance Publique - Hôpitaux de Paris (AP-HP), Centre d'Investigation Clinique (CIC-1901), Pharmacology Department, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
256
|
Mirzayev I, Gündüz AK, Mirzayeva L, Ceyhan K. Extended survival in a case of metastatic choroidal melanoma with immunotherapy. Anticancer Drugs 2024; 35:974-978. [PMID: 39079167 DOI: 10.1097/cad.0000000000001650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2024]
Abstract
Uveal melanoma is the most common intraocular malignancy in adults. Despite advances in local treatments, approximately 50% of all cases eventually die from metastatic disease. In cases with metastasis, 2- and 5-year survival rates are approximately 10% and <1%, respectively. Advances in molecular biology have led to the identification of a number of promising drugs including immune checkpoint inhibitors (ICIs). Ipilimumab and nivolumab are ICIs targeting the cytotoxic T-lymphocyte-associated antigen-4 and the programmed-cell death protein-1, respectively. Herein, we present a case of choroidal melanoma having liver metastasis treated with nivolumab and ipilimumab and transarterial radioembolization, achieving a 3-year survival.
Collapse
Affiliation(s)
- Ibadulla Mirzayev
- Department of Ophthalmology, Ankara University Faculty of Medicine
- Halil Şivgin Çubuk Devlet Hastanesi
| | - Ahmet Kaan Gündüz
- Department of Ophthalmology, Ankara University Faculty of Medicine
- Private Eye Clinic
| | - Leyla Mirzayeva
- Department of Radiology, Gazi University Faculty of Medicine
| | - Koray Ceyhan
- Department of Pathology, Ankara University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
257
|
Márquez-Rodas I, Muñoz Couselo E, Rodríguez Moreno JF, Arance Fernández AM, Berciano Guerrero MÁ, Campos Balea B, de la Cruz Merino L, Espinosa Arranz E, García Castaño A, Berrocal Jaime A. SEOM-GEM clinical guidelines for cutaneous melanoma (2023). Clin Transl Oncol 2024; 26:2841-2855. [PMID: 38748192 PMCID: PMC11467041 DOI: 10.1007/s12094-024-03497-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 10/11/2024]
Abstract
Cutaneous melanoma incidence is rising. Early diagnosis and treatment administration are key for increasing the chances of survival. For patients with locoregional advanced melanoma that can be treated with complete resection, adjuvant-and more recently neoadjuvant-with targeted therapy-BRAF and MEK inhibitors-and immunotherapy-anti-PD-1-based therapies-offer opportunities to reduce the risk of relapse and distant metastases. For patients with advanced disease not amenable to radical treatment, these treatments offer an unprecedented increase in overall survival. A group of medical oncologists from the Spanish Society of Medical Oncology (SEOM) and Spanish Multidisciplinary Melanoma Group (GEM) has designed these guidelines, based on a thorough review of the best evidence available. The following guidelines try to cover all the aspects from the diagnosis-clinical, pathological, and molecular-staging, risk stratification, adjuvant therapy, advanced disease therapy, and survivor follow-up, including special situations, such as brain metastases, refractory disease, and treatment sequencing. We aim help clinicians in the decision-making process.
Collapse
Affiliation(s)
| | - Eva Muñoz Couselo
- Hospital Vall d'Hebron & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | | | | | - Luis de la Cruz Merino
- Cancer Immunotherapy, Biomedicine Institute of Seville (IBIS)/CSIC, Clinical Oncology Department, University Hospital Virgen Macarena and School of Medicine, University of Seville, Seville, Spain
| | | | | | | |
Collapse
|
258
|
Woodford R, McKeown J, Hoeijmakers LL, Mangana J, Dimitriou F, Allayous C, Zaman F, Aya F, Marsiglio J, Goodman R, Rayson V, Placzke J, Kessels J, Ramalyte E, Haque W, Wilson I, Trojaniello C, Benannoune N, Roberts-Thomson R, Robert C, Blank CU, Dummer R, Lebbe C, Haydon A, Arance A, Hu-Lieskovan S, Johnson DB, Mcarthur GA, Rutkowski P, Neyns B, Sullivan RJ, Weber J, Carlino MS, Ascierto PA, Lo S, Long GV, Menzies AM. Nature and management of melanoma recurrences following adjuvant anti-PD-1 based therapy. Eur J Cancer 2024; 212:115055. [PMID: 39366209 DOI: 10.1016/j.ejca.2024.115055] [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: 07/02/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024]
Abstract
INTRODUCTION Approximately 50 % of resected stage II-IV melanoma patients develop recurrent disease by 5 years despite adjuvant anti-PD-1 therapy. Data to define best management of recurrences is lacking. METHODS This was a multicentre, international, retrospective cohort study. Patients with resected stage II-IV melanoma who commenced adjuvant anti-PD-1-based therapy before January 2022 and later recurred were identified. Data on demographics, disease characteristics, recurrence patterns, management and outcomes were collected. RESULTS 711 patients from 17 sites were included. Median age was 60 [range 16-92], 64 % were male, 2 % stage II, 91 % were stage III, 7 % stage IV. Median time to recurrence was 6.2 months (0-68.5) and median follow up time from recurrence was 19.8 months (range 0.2-73.1). 63 % recurred on anti-PD-1 therapy, 36 % off therapy [3 % < 6 months, 33 % > 6 months]. Initial recurrences were locoregional (LR) alone in 44 %, distant alone (DR) in 43 %, and 11 % in both sites. LR recurrences were managed with local therapy, alone (62 %) or with "second adjuvant" anti-PD-1 (14 %) or BRAF/MEK therapy (23 %); 12 m RFS2 was 25 %, 29 % and 69 % respectively (p = 0.0045). Definitive systemic therapy at first recurrence was given in 16 % LR and 86 % DR, with best outcomes for anti-CTLA4 + anti-PD-1 and trial combinations (24 m PFS 63 % and 69 %, respectively). The 24 m OS for the entire cohort was 65 %. CONCLUSION Most recurrences following adjuvant anti-PD-1 based therapy occur early and while still on drug. Outcomes are poor, regardless of site, timing of recurrence, and subsequent treatment.
Collapse
Affiliation(s)
- Rachel Woodford
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia
| | - Janet McKeown
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia
| | - Lotte L Hoeijmakers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, Amsterdam, The Netherlands
| | - Johanna Mangana
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Ramistrasse 100, Zurich, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Ramistrasse 100, Zurich, Switzerland
| | - Clara Allayous
- APHP Hospital St-Louis, Dermatology Department, DMU ICARE, Paris, France
| | - Farzana Zaman
- Department of Medical Oncology, The Alfred Hospital, 55 Commercial Road, Melbourne, Victoria, Australia
| | - Francisco Aya
- Hospital Clinic Barcelona, Calle de Villaroel 170, Barcelona, Spain
| | - John Marsiglio
- Huntsman Cancer Institute, University of Utah School of Medicine, UT, USA
| | - Rachel Goodman
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Victoria Rayson
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia
| | - Joanna Placzke
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skowska-Curie Research Institute of Oncology, Masovian Voivodeship, Warsaw, Poland
| | - Jolien Kessels
- Department of Medical Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Belgium
| | | | - Waqas Haque
- Department of Medicine, New York University Grossman School of Medicine Langone, Manhattan, NY, USA
| | - Isabella Wilson
- Department of Medical Oncology, Westmead and Blacktown Hospitals, NSW, Australia
| | - Claudia Trojaniello
- Unit of Melanoma Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Naima Benannoune
- Dermatology Unit, Gustave-Roussy Paris Saclay University, 39 Rue Camille Desmoulins, Villejuif, France
| | | | - Caroline Robert
- Dermatology Unit, Gustave-Roussy Paris Saclay University, 39 Rue Camille Desmoulins, Villejuif, France
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, Amsterdam, The Netherlands
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Ramistrasse 100, Zurich, Switzerland
| | - Celeste Lebbe
- APHP Hospital St-Louis, Dermatology Department, DMU ICARE, Paris, France; Universite de Paris Cité, 45 Rue des Saint-Pères, Paris, France
| | - Andrew Haydon
- Department of Medical Oncology, The Alfred Hospital, 55 Commercial Road, Melbourne, Victoria, Australia
| | - Ana Arance
- Hospital Clinic Barcelona, Calle de Villaroel 170, Barcelona, Spain
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah School of Medicine, UT, USA
| | | | - Grant A Mcarthur
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skowska-Curie Research Institute of Oncology, Masovian Voivodeship, Warsaw, Poland
| | - Bart Neyns
- Department of Medical Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Belgium
| | | | - Jeffrey Weber
- Department of Medicine, New York University Grossman School of Medicine Langone, Manhattan, NY, USA
| | - Matteo S Carlino
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia; Department of Medical Oncology, Westmead and Blacktown Hospitals, NSW, Australia
| | - Paolo A Ascierto
- Unit of Melanoma Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Serigne Lo
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, North Sydney, Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, North Sydney, Sydney, NSW, Australia.
| |
Collapse
|
259
|
Fox AC, Blazeck J. Applying metabolic control strategies to engineered T cell cancer therapies. Metab Eng 2024; 86:250-261. [PMID: 39490640 PMCID: PMC11611646 DOI: 10.1016/j.ymben.2024.10.009] [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: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an engineered immunotherapy that express synthetic receptors to recognize and kill cancer cells. Despite their success in treating hematologic cancers, CAR T cells have limited efficacy against solid tumors, in part due to the altered immunometabolic profile within the tumor environment, which hinders T cell proliferation, infiltration, and anti-tumor activity. For instance, CAR T cells must compete for essential nutrients within tumors, while resisting the impacts of immunosuppressive metabolic byproducts. In this review, we will describe the altered metabolic features within solid tumors that contribute to immunosuppression of CAR T cells. We'll discuss how overexpression of key metabolic enzymes can enhance the ability of CAR T cells to resist corresponding tumoral metabolic changes or even revert the metabolic profile of a tumor to a less inhibitory state. In addition, metabolic remodeling is intrinsically linked to T cell activity, differentiation, and function, such that metabolic engineering strategies can also promote establishment of more or less efficacious CAR T cell phenotypes. Overall, we will show how applying metabolic engineering strategies holds significant promise in improving CAR T cells for the treatment of solid tumors.
Collapse
Affiliation(s)
- Andrea C Fox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA.
| |
Collapse
|
260
|
Aziz H, Kwon YIC, Park AMG, Lai A, Lee KYC, Zhang D, Kwon Y, Pawlik TM. Recent advancements in management for noncolorectal, nonneuroendocrine hepatic metastases. J Gastrointest Surg 2024; 28:1922-1932. [PMID: 39154708 DOI: 10.1016/j.gassur.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Owing to the heterogeneity of underlying primary tumors, noncolorectal, nonneuroendocrine metastases to the liver (NCNNMLs), although relatively rare, pose major challenges to treatment and long-term management. Despite being considered the gold standard for colorectal cancer liver metastases, the role of surgical resection for NCNNML remains controversial. Furthermore, advancements in locoregional treatment modalities, such as ablation and various chemotherapeutic modalities, have contributed to the treatment of patients with NCNNML. METHODS This was a comprehensive review of literature that used Medline/PubMed, Google Scholar, the Cochrane Library, and the Web of Science, which were accessed between 2014 and 2024. RESULTS NCNNMLs are rare tumor entities with varied presentation and outcomes. A multidisciplinary approach, which includes chemotherapy, surgery, and interventional radiologic techniques, can be implemented with good results. CONCLUSION Given the complex nature of NCNNML, its management should be highly individualized and multidisciplinary. Locoregional treatments, such as surgical resection and/or ablation, may be more appropriate for select patients and should be offered as a viable therapeutic option for a subset of individuals.
Collapse
Affiliation(s)
- Hassan Aziz
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Ye In Christopher Kwon
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Andrew Min-Gi Park
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Kerry Yi Chen Lee
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Dean Zhang
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yeseo Kwon
- Department of Surgery, School of Medicine, Tufts University, Boston, MA, United States
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| |
Collapse
|
261
|
Tschanz J, Khan R, Bruera E. Case report: Poor prognosis or poor prognostication? Palliat Support Care 2024; 23:e11. [PMID: 39478369 DOI: 10.1017/s1478951524001512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
OBJECTIVES This case highlights the limitations of current prognostication and communication in clinical practice. METHODS We report a case of a 50 year old patient with metastatic melanoma following admission to intensive care unit and later transferred to palliative care unit for end-of-life care. RESULTS The patient had clinical improvement despite signs of predictors of death and was later transferred back to care of oncology team. SIGNIFICANCE OF RESULTS Physicians frequently overestimate or underestimate survival time which can be distressing to patients and families. There is need for further research to improve the accuracy of these tools for the sake of our patients and their families.
Collapse
Affiliation(s)
- Jacqueline Tschanz
- Department of Palliative Care, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Rida Khan
- Department of Palliative Care, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Eduardo Bruera
- Department of Palliative Care, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| |
Collapse
|
262
|
Bridges K, Pizzurro GA, Baysoy A, Baskaran JP, Xu Z, Mathew V, Tripple V, LaPorte M, Park K, Damsky W, Kluger H, Fan R, Kaech SM, Bosenberg MW, Miller-Jensen K. Mapping intratumoral myeloid-T cell interactomes at single-cell resolution reveals targets for overcoming checkpoint inhibitor resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620093. [PMID: 39554094 PMCID: PMC11565996 DOI: 10.1101/2024.10.28.620093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Effective cancer immunotherapies restore anti-tumor immunity by rewiring cell-cell communication. Treatment-induced changes in communication can be inferred from single-cell RNA-sequencing (scRNA-seq) data, but current methods do not effectively manage heterogeneity within cell types. Here we developed a computational approach to efficiently analyze scRNA-seq-derived, single-cell-resolved cell-cell interactomes, which we applied to determine how agonistic CD40 (CD40ag) alters immune cell crosstalk alone, across tumor models, and in combination with immune checkpoint blockade (ICB). Our analyses suggested that CD40ag improves responses to ICB by targeting both immuno-stimulatory and immunosuppressive macrophage subsets communicating with T cells, and we experimentally validated a spatial basis for these subsets with immunofluorescence and spatial transcriptomics. Moreover, treatment with CD40ag and ICB established coordinated myeloid-T cell interaction hubs that are critical for reestablishing antitumor immunity. Our work advances the biological significance of hypotheses generated from scRNA-seq-derived cell-cell interactomes and supports the clinical translation of myeloid-targeted therapies for ICB-resistant tumors.
Collapse
Affiliation(s)
- Kate Bridges
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Present address: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Alev Baysoy
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Janani P. Baskaran
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Ziyan Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Varsha Mathew
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael LaPorte
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Koonam Park
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Harriet Kluger
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Marcus W. Bosenberg
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Systems Biology Institute, Yale University, New Haven, CT 06511, USA
- Lead contact
| |
Collapse
|
263
|
Huang AY, Burke KP, Porter R, Meiger L, Fatouros P, Yang J, Robitschek E, Vokes N, Ricker C, Rosado V, Tarantino G, Chen J, Aprati TJ, Glettig MC, He Y, Wang C, Fu D, Ho LL, Galani K, Freeman GJ, Buchbinder EI, Stephen Hodi F, Kellis M, Boland GM, Sharpe AH, Liu D. Stratified analysis identifies HIF-2 α as a therapeutic target for highly immune-infiltrated melanomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620300. [PMID: 39554029 PMCID: PMC11565796 DOI: 10.1101/2024.10.29.620300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
While immune-checkpoint blockade (ICB) has revolutionized treatment of metastatic melanoma over the last decade, the identification of broadly applicable robust biomarkers has been challenging, driven in large part by the heterogeneity of ICB regimens and patient and tumor characteristics. To disentangle these features, we performed a standardized meta-analysis of eight cohorts of patients treated with anti-PD-1 (n=290), anti-CTLA-4 (n=175), and combination anti-PD-1/anti-CTLA-4 (n=51) with RNA sequencing of pre-treatment tumor and clinical annotations. Stratifying by immune-high vs -low tumors, we found that surprisingly, high immune infiltrate was a biomarker for response to combination ICB, but not anti-PD-1 alone. Additionally, hypoxia-related signatures were associated with non-response to anti-PD-1, but only amongst immune infiltrate-high melanomas. In a cohort of scRNA-seq of patients with metastatic melanoma, hypoxia also correlated with immunosuppression and changes in tumor-stromal communication in the tumor microenvironment (TME). Clinically actionable targets of hypoxia signaling were also uniquely expressed across different cell types. We focused on one such target, HIF-2α, which was specifically upregulated in endothelial cells and fibroblasts but not in immune cells or tumor cells. HIF-2α inhibition, in combination with anti-PD-1, enhanced tumor growth control in pre-clinical models, but only in a more immune-infiltrated melanoma model. Our work demonstrates how careful stratification by clinical and molecular characteristics can be leveraged to derive meaningful biological insights and lead to the rational discovery of novel clinical targets for combination therapy.
Collapse
Affiliation(s)
- Amy Y Huang
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Massachusetts Institute of Technology, Cambridge, USA
| | - Kelly P Burke
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryan Porter
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lynn Meiger
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Fatouros
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jiekun Yang
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, USA
- Rutgers University, New Brunswick, NJ, USA
| | - Emily Robitschek
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Natalie Vokes
- University of Texas MD Anderson Cancer Center, Houston, USA
| | - Cora Ricker
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Valeria Rosado
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Giuseppe Tarantino
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jiajia Chen
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tyler J Aprati
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc C Glettig
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- ETH Zürich, Zurich, Switzerland
| | - Yiwen He
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cassia Wang
- Massachusetts Institute of Technology, Cambridge, USA
| | - Doris Fu
- Massachusetts Institute of Technology, Cambridge, USA
| | - Li-Lun Ho
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, USA
| | - Kyriakitsa Galani
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, USA
| | - Gordon J Freeman
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - F Stephen Hodi
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Manolis Kellis
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, USA
| | - Genevieve M Boland
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Arlene H Sharpe
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Liu
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
264
|
Watts T, Jennings S, Anstey S, Roche D. Supporting People Treated with Immune Checkpoint Inhibitors: A Qualitative Study Exploring Oncology Healthcare Professionals' Experiences. Semin Oncol Nurs 2024:151745. [PMID: 39482207 DOI: 10.1016/j.soncn.2024.151745] [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: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Immune checkpoint inhibitors have recently developed successfully in treatment for several advanced cancers, including advanced renal cancer, where options have previously been limited. However, while some are able to tolerate these treatments, others may experience unpredictable and sometimes severe immune-related adverse events. Oncology health care professionals have vital roles in optimizing safety and supporting positive outcomes for people receiving these treatments. This study aimed to better understand these professionals' experiences of supporting people receiving immune checkpoint inhibitors. METHODS A qualitative exploratory methodology was adopted using semi-structured interviews with 18 purposively sampled senior oncology health professionals, including 12 nurses, who had experience caring for people being treated with checkpoint inhibitors. Data were collected between June and September 2020, transcribed verbatim, and analyzed using reflexive thematic analysis. RESULTS The analysis identified three main themes: First, participants were positive about the potential benefits that checkpoint inhibitors afforded patients, balanced against challenges associated with ambiguities of the treatments and potential impact on existing workloads. Secondly, participants identified the importance of proactive patient monitoring for early detection and reporting of adverse events. Participants highlighted potential challenges if these events went undetected, particularly in the context of the expectation for patient recognition and prompt reporting. Finally, participants identified the need for continual enhancement of health professionals' knowledge and understanding of immunotherapy, supported by the prioritizing of formal immunotherapy education. CONCLUSIONS Whilst immune checkpoint inhibitors offer the possibility for improved disease outcomes, this is balanced against uncertainties regarding potentially unpredictable, often complex, adverse treatment events. This study shows that nurses have vital roles in supporting people receiving these treatments. IMPLICATIONS FOR NURSING PRACTICE Effective care and treatment management for people receiving checkpoint inhibitors require nurses' support through their expert knowledge of immunotherapy and their skills for appropriate coordination and organization of cross-boundary care.
Collapse
Affiliation(s)
- Tessa Watts
- Reader, School of Healthcare Sciences, Cardiff University, Cardiff, Wales.
| | - Stephen Jennings
- Lecturer, Bristol Medical School, Bristol University, Cardiff, Wales
| | - Sally Anstey
- Reader Emirata, School of Healthcare Sciences, Cardiff University, Cardiff, Wales
| | - Dominic Roche
- Senior Lecturer, School of Healthcare Sciences, Cardiff University, Cardiff, Wales
| |
Collapse
|
265
|
Li AY, Bu J, Xiao HN, Zhao ZY, Zhang JL, Yu B, Li H, Li JP, Xiao T. Two-step consensus clustering approach to immune cell infiltration: An integrated exploration and validation of prognostic and immune implications in sarcomas. Heliyon 2024; 10:e38253. [PMID: 39492897 PMCID: PMC11531637 DOI: 10.1016/j.heliyon.2024.e38253] [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: 03/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 11/05/2024] Open
Abstract
To conduct a comprehensive investigation of the sarcoma immune cell infiltration (ImmCI) patterns and tumoral microenvironment (TME). We utilized transcriptomic, clinical, and mutation data of sarcoma patients (training cohort) obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) server. Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithms were applied to decipher the immune cell infiltration landscape and TME profiles of sarcomas. An unsupervised clustering method was utilized for classifying ImmCI clusters (initial clustering) and ImmCI-based differentially expressed gene-driven clusters (secondary clustering). Mortality rates and immune checkpoint gene levels was analyzed among the identified clusters. We calculated the ImmCI score through principal component analysis. The tumor immune dysfunction evaluation (TIDE) score was also employed to quantify immunotherapy efficacy between two ImmCI score groups. We further validated the biomarkers for ImmCI and gene-driven clusters via experimental verification and the accuracy of the ImmCI score in predicting survival outcomes and immunotherapy efficacy by external validation cohorts (testing cohort). We demonstrated that ImmCI cluster A and gene-driven cluster A, were beneficial prognostic biomarkers and indicators of immune checkpoint blockade response in sarcomas via in-silico and laboratory experiments. Additionally, the ImmCI score exhibited independent prognostic significance and was predictive of immunotherapy response. Our research underscores the clinical significance of ImmCI scores in identifying sarcoma patients likely to respond to immunotherapy.
Collapse
Affiliation(s)
- Ao-Yu Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jie Bu
- Department of Orthopedics, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui-Ni Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Zi-Yue Zhao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jia-Lin Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Bin Yu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jin-Ping Li
- Department of Orthopedics, Changsha Central Hospital, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| |
Collapse
|
266
|
Cao WH, Zhang YQ, Li XX, Zhang ZY, Li MH. Advances in immunotherapy for hepatitis B virus associated hepatocellular carcinoma patients. World J Hepatol 2024; 16:1158-1168. [PMID: 39474576 PMCID: PMC11514615 DOI: 10.4254/wjh.v16.i10.1158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024] Open
Abstract
Hepatitis B virus (HBV) infection plays an important role in the occurrence and development of hepatocellular carcinoma (HCC), and the rate of HBV infection in liver cancer patients in China is as high as 92.05%. Due to long-term exposure to chronic antigens from the gut, the liver needs to maintain a certain level of immune tolerance, both to avoid severe inflammation caused by non-pathogenic antigens and to maintain the possibility of rapid and violent responses to infection and tumors. Therefore, HBV infection interacts with the tumor microenvironment (TME) through a highly complex and intertwined signaling pathway, which results in a special TME in HCC. Due to changes in the TME, tumor cells can evade immune surveillance by inhibiting tumor-specific T cell function through cytotoxic T-lymphocy-associated protein-4 (CTLA-4) and programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1). Interferons, as a class of immune factors with strong biological activity, can improve the TME of HBV-HCC through various pathways. In recent years, the systematic treatment of HCC has gradually come out of the dilemma. In addition to the continuous emergence of new multi-target anti-vascular tyrosine kinase inhibitor drugs, immune checkpoint inhibitors have opened up a new avenue for the systematic treatment of HCC. At present, immunotherapy based on PD-1/L1 inhibitors has gradually become a new direction of systematic treatment for HCC, and the disease characteristics of patients included in global clinical studies are different from those of Chinese patients. Therefore, whether a group of HCC patients with HBV background and poor prognosis in China can also benefit from immunotherapy is an issue of wide concern. This review aims to elucidate the advances of immunotherapy for HBV related HCC patients with regard to: (1) Immunotherapy based on interferons; (2) Immunotherapy based on PD-1/L1 inhibitors; (3) Immunotherapy based on CTLA4 inhibitors; (4) Adoptive cell transfer; (5) Combination immunotherapy strategy; and (6) Shortcomings of immunotherapy.
Collapse
Affiliation(s)
- Wei-Hua Cao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ya-Qin Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xin-Xin Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Zi-Yu Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ming-Hui Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Peking University Ditan Teaching Hospital, Beijing 100015, China
| |
Collapse
|
267
|
Cao WH, Zhang YQ, Li XX, Zhang ZY, Li MH. Advances in immunotherapy for hepatitis B virus associated hepatocellular carcinoma patients. World J Hepatol 2024; 16:1338-1348. [DOI: 10.4254/wjh.v16.i10.1338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 11/22/2024] Open
Abstract
Hepatitis B virus (HBV) infection plays an important role in the occurrence and development of hepatocellular carcinoma (HCC), and the rate of HBV infection in liver cancer patients in China is as high as 92.05%. Due to long-term exposure to chronic antigens from the gut, the liver needs to maintain a certain level of immune tolerance, both to avoid severe inflammation caused by non-pathogenic antigens and to maintain the possibility of rapid and violent responses to infection and tumors. Therefore, HBV infection interacts with the tumor microenvironment (TME) through a highly complex and intertwined signaling pathway, which results in a special TME in HCC. Due to changes in the TME, tumor cells can evade immune surveillance by inhibiting tumor-specific T cell function through cytotoxic T-lymphocy-associated protein-4 (CTLA-4) and programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1). Interferons, as a class of immune factors with strong biological activity, can improve the TME of HBV-HCC through various pathways. In recent years, the systematic treatment of HCC has gradually come out of the dilemma. In addition to the continuous emergence of new multi-target anti-vascular tyrosine kinase inhibitor drugs, immune checkpoint inhibitors have opened up a new avenue for the systematic treatment of HCC. At present, immunotherapy based on PD-1/L1 inhibitors has gradually become a new direction of systematic treatment for HCC, and the disease characteristics of patients included in global clinical studies are different from those of Chinese patients. Therefore, whether a group of HCC patients with HBV background and poor prognosis in China can also benefit from immunotherapy is an issue of wide concern. This review aims to elucidate the advances of immunotherapy for HBV related HCC patients with regard to: (1) Immunotherapy based on interferons; (2) Immunotherapy based on PD-1/L1 inhibitors; (3) Immunotherapy based on CTLA4 inhibitors; (4) Adoptive cell transfer; (5) Combination immunotherapy strategy; and (6) Shortcomings of immunotherapy.
Collapse
Affiliation(s)
- Wei-Hua Cao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ya-Qin Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xin-Xin Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Zi-Yu Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ming-Hui Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Peking University Ditan Teaching Hospital, Beijing 100015, China
| |
Collapse
|
268
|
Liu H, Lu Y, Zong J, Zhang B, Li X, Qi H, Yu T, Li Y. Engineering dendritic cell biomimetic membrane as a delivery system for tumor targeted therapy. J Nanobiotechnology 2024; 22:663. [PMID: 39465376 PMCID: PMC11520105 DOI: 10.1186/s12951-024-02913-7] [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: 05/17/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024] Open
Abstract
Targeted immunotherapies make substantial strides in clinical cancer care due to their ability to counteract the tumor's capacity to suppress immune responses. Advances in biomimetic technology with minimally immunogenic and highly targeted, are addressing issues of targeted drug delivery and disrupting the tumor's immunosuppressive environment to trigger immune activation. Specifically, the use of dendritic cell (DC) membranes to coat nanoparticles ensures targeted delivery due to DC's unique ability to activate naive T cells, spotlighting their role in immunotherapy aimed at disrupting the tumor microenvironment. The potential of DC's biomimetic membrane to mediate immune activation and target tumors is gaining momentum, enhancing the effectiveness of cancer treatments in conjunction with other immune responses. This review delves into the methodologies behind crafting DC membranes and the fusion of dendritic and tumor cell membranes for encapsulating therapeutic nanoparticles. It explores their applications and recent advancements in combating cancer, offering an all-encompassing perspective on DC biomimetic nanosystems, immunotherapy driven by antigen presentation, and the collaborative efforts of drug delivery in chemotherapy and photodynamic therapies. Current evidence shows promise in augmenting combined therapeutic approaches for cancer treatment and holds translational potential for various cancer treatments in a clinical setting.
Collapse
Affiliation(s)
- Huiyang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Yiming Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China.
| |
Collapse
|
269
|
Misiąg P, Molik K, Kisielewska M, Typek P, Skowron I, Karwowska A, Kuźnicki J, Wojno A, Ekiert M, Choromańska A. Amelanotic Melanoma-Biochemical and Molecular Induction Pathways. Int J Mol Sci 2024; 25:11502. [PMID: 39519055 PMCID: PMC11546312 DOI: 10.3390/ijms252111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Amelanotic melanoma (AM) is a subtype of hypomelanotic or completely amelanotic melanoma. AM is a rare subtype of melanoma that exhibits a higher recurrence rate and aggressiveness as well as worse surveillance than typical melanoma. AM shows a dysregulation of melanin production, cell cycle control, and apoptosis pathways. Knowing these pathways has an application in medicine due to targeted therapies based on the inhibiting elements of the abovementioned pathways. Therefore, we summarized and discussed AM biochemical and molecular induction pathways and personalized medicine approaches, clinical management, and future directions due to the fact that AM is relatively rare. AM is commonly misdiagnosed. Hence, the role of biomarkers is becoming significant. Nonetheless, there is a shortage of biomarkers specific to AM. BRAF, NRAS, and c-KIT genes are the main targets of therapy. However, the role of BRAF and KIT in AM varied among studies. BRAF inhibitors combined with MAK inhibitors demonstrate better results. Immune checkpoint inhibitors targeting CTLA-4 combined with a programmed death receptor 1 (PD-1) show better outcomes than separately. Fecal microbiota transplantation may overcome resistance to immune checkpoint therapy of AM. Immune-modulatory vaccines against indoleamine 2,3-dioxygenase (IDO) and PD ligand (PD-L1) combined with nivolumab may be efficient in melanoma treatment.
Collapse
Affiliation(s)
- Piotr Misiąg
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Monika Kisielewska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Paulina Typek
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Anna Karwowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Jacek Kuźnicki
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Aleksandra Wojno
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (P.M.); (K.M.); (M.K.); (P.T.); (I.S.); (A.K.); (J.K.); (A.W.)
- Students Scientific Group No. 148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Marcin Ekiert
- Department of Oncology, Wroclaw Medical University, pl. L. Hirszfelda 12, 53-413 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
270
|
Miao W, Jain V, Han M, Jin YJ, Beasley GM, Starczysnowski DT, Gregory SG, Zhang JY. Inhibition of UBE2N in regulatory T-cells boosts immunity against cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619436. [PMID: 39484562 PMCID: PMC11526935 DOI: 10.1101/2024.10.22.619436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Regulatory T (Treg) cells prevent autoimmunity and facilitate cancer immune evasion. Depletion of Tregs is a promising cancer therapy, but risks of autoimmune reactions hamper its clinical translation. Here, we demonstrate that temporally induced deletion of Ube2n in Tregs (Ube2n Treg-KO ) of adult mice results in a robust expansion and activation of cytotoxic CD8 + T-cells in response to cancer cell challenges, producing a long-lasting survival benefit without autoimmune complications. The anti-tumor effect persists following adoptive T-cell transfer to T-cell-deficient Rag1-knockout mice. Single-cell transcriptomic analysis revealed that UBE2N deletion shifted immunosuppressive Tregs to effector-like T-cells. This shift is characterized by the downregulation of c-Myc target genes, resembling that observed in tumor-infiltrating Tregs of melanoma patients. Further analyses confirm that UBE2N maintains c-Myc protein stability via suppression of K48-Ubiquitin-mediated proteasomal degradation. Taken together, our studies uncover a hitherto unexplored and potentially druggable UBE2N/c-Myc signaling axis to eradicate Treg-enabled cancer immune escape.
Collapse
|
271
|
Huang H, Tong QS, Chen Y, Liu XY, Liu R, Shen S, Du JZ, Wang J. PAMAM-Based Polymeric Immunogenic Cell Death Inducer To Potentiate Cancer Immunotherapy. J Am Chem Soc 2024; 146:29189-29198. [PMID: 39387453 DOI: 10.1021/jacs.4c11636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Immunogenic cell death (ICD) has been widely employed to potentiate cancer immunotherapy due to its capability to activate the anticancer immune response. Although various ICD inducers have been described, the development of synthetic materials with intrinsic ICD-inducing competency has rarely been reported. Herein, we identify a derivative of the fourth generation polyamidoamine (PAMAM) modified with multiple seven-membered heterocyclic rings, G4P-C7A, as a robust ICD inducer. G4P-C7A evokes characteristic release of damage-associated molecular patterns in tumor cells and induces efficient dendritic cell maturation. Mechanistic studies suggest that G4P-C7A can selectively accumulate in the endoplasmic reticulum and mitochondria to generate reactive oxygen species. G4P-C7A-treated tumor cells can work as potent vaccines to protect against secondary tumor implantation. Either local or systemic injection of G4P-C7A alone can effectively inhibit tumor growth by eliciting robust antitumor immune response. The combination of G4P-C7A with immunotherapeutic antibodies such as anti-PD1 (aPD-1) and anti-CD47 (aCD47) further potentiates the antitumor effect in either CT26 or 4T1 tumor model. This study offers a simple but effective strategy to induce ICD to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Hua Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Qi-Song Tong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yang Chen
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xiao-Yue Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
272
|
Jiang Q, Chen X, Wu J, Wei S, Tao R. Oral Lichenoid lesions induced by programmed cell death protein 1 and cytotoxic T-lymphocyte-associated protein 4 bispecific antibody: a case report. BMC Oral Health 2024; 24:1240. [PMID: 39425127 PMCID: PMC11490079 DOI: 10.1186/s12903-024-05036-5] [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: 08/15/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Cadonilimab is the first approved dual immune checkpoint inhibitor targeting programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), currently utilized for the treatment of various solid tumors. Oral mucosal adverse reactions, such as oral lichenoid lesions, represent one of the most prevalent immune-related adverse events associated with immune checkpoint antibodies. However, reports detailing oral side effects specifically linked to Cadonilimab are lacking. Documenting these side effects is essential to alert oncologists and stomatologists, facilitating timely intervention for affected patients. CASE PRESENTATION We present a case involving a 35-year-old male patient diagnosed with hepatocellular carcinoma who received Cadonilimab following hepatectomy and subsequently developed extensive oral lichenoid lesions along with mucosal erosion at 13-14 weeks post-treatment initiation. A biopsy was conducted revealing immunohistochemical findings of CD3+, CD4+, CD8+, CD20 + lymphocytes, CD68 + macrophages, and α-SMA + myofibroblasts infiltrating the tissue of the oral lichenoid lesions. The patient's oral lesions improved after administration of systemic and local glucocorticoid therapy alongside cessation of Cadonilimab treatment. CONCLUSION This report marks the first documented instance of an oral adverse effect associated with Cadonilimab use. It underscores that administration of this agent may lead to significant lichenoid lesions and erosions within the oral cavity-an issue warranting increased vigilance from both oncologists and stomatologists.
Collapse
MESH Headings
- Humans
- Male
- Adult
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/therapeutic use
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- CTLA-4 Antigen/antagonists & inhibitors
- Lichen Planus, Oral/pathology
- Lichen Planus, Oral/drug therapy
- Lichen Planus, Oral/chemically induced
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/therapeutic use
- Lichenoid Eruptions/chemically induced
- Lichenoid Eruptions/pathology
Collapse
Affiliation(s)
- Qiaozhi Jiang
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, P. R. China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, P. R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, P. R. China
| | - Xinyu Chen
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, P. R. China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, P. R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, P. R. China
| | - Jiaxuan Wu
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, P. R. China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, P. R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, P. R. China
| | - Shanni Wei
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, P. R. China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, P. R. China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, P. R. China
| | - Renchuan Tao
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, P. R. China.
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, P. R. China.
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Universities and Colleges Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, Guangxi, P. R. China.
| |
Collapse
|
273
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
274
|
Zhen J, Chen L, Wang H, Li D, Lai M, Ding Y, Yang Y, Li J, Wen X, Cai L, Zhang X. Intrathecal anti-PD-1 treatment in metastatic melanoma patients with leptomeningeal disease (LMD): real-world data and evidence. J Neurooncol 2024:10.1007/s11060-024-04843-8. [PMID: 39422814 DOI: 10.1007/s11060-024-04843-8] [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: 02/21/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Leptomeningeal disease (LMD) is a severe complication of melanoma with a very poor prognosis. Despite improved treatment strategies and prolonged survival, the incidence of LMD has increased over the past decade. This real-world study aims to evaluate the efficacy and safety of intrathecal anti-PD-1 treatment in melanoma patients with LMD. METHODS Melanoma patients with LMD diagnosed by magnetic resonance imaging (MRI) and/or cerebrospinal fluid (CSF) cytology were treated with intrathecal infusions of nivolumab 20 mg once every 2 weeks (n = 5) or pembrolizumab 20 mg once every 3 weeks (n = 3), alongside systemic therapy. Patients received a median of 5.5 treatment cycles (range 2-9). Efficacy and safety analyses were performed on all treated patients. RESULTS From June 2022 to February 2023, eight patients were treated, including four with cutaneous melanoma, two with acral melanoma, and two with primary leptomeningeal melanoma. All patients exhibited linear or small nodular enhancement of the leptomeninges on MRI. Four patients had concurrent parenchymal brain metastases. Tumor cells were identified in six patients by CSF cytology, and two patients underwent leptomeningeal biopsy for pathological diagnosis. According to the RANO-LM criteria, five patients responded to treatment with symptom improvement and reduction or disappearance of linear enhancement on MRI, while three patients developed progressive disease. With a median follow-up of 20.7 weeks (range 8.1-45.3 weeks), the median OS and median intracranial progression-free survival (IPFS) for intrathecal anti-PD-1 treatment were 21.1 and 16.1 weeks, respectively. All treatment-related adverse events were grade 1-2, including headache (grade 1, n = 1; grade 2, n = 2) and low back pain (grade 1, n = 1). CONCLUSIONS In this real-world study, intrathecal anti-PD-1 treatment demonstrated potential clinical benefits and was well tolerated in metastatic melanoma patients with LMD.
Collapse
Affiliation(s)
- Junjie Zhen
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, 510060, PR China
| | - Linbin Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Hui Wang
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, 510060, PR China
| | - Dandan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Mingyao Lai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, 510060, PR China
| | - Ya Ding
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Yanying Yang
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, 510060, PR China
| | - Jingjing Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xizhi Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Linbo Cai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, Guangzhou, 510060, PR China.
| | - Xiaoshi Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| |
Collapse
|
275
|
Ascic E, Åkerström F, Nair MS, Rosa A, Kurochkin I, Zimmermannova O, Catena X, Rotankova N, Veser C, Rudnik M, Ballocci T, Schärer T, Huang X, de Rosa Torres M, Renaud E, Santiago MV, Met Ö, Askmyr D, Lindstedt M, Greiff L, Ligeon LA, Agarkova I, Svane IM, Pires CF, Rosa FF, Pereira CF. In vivo dendritic cell reprogramming for cancer immunotherapy. Science 2024; 386:eadn9083. [PMID: 39236156 PMCID: PMC7616765 DOI: 10.1126/science.adn9083] [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: 02/29/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Immunotherapy can lead to long-term survival for some cancer patients, yet generalized success has been hampered by insufficient antigen presentation and exclusion of immunogenic cells from the tumor microenvironment. Here, we developed an approach to reprogram tumor cells in vivo by adenoviral delivery of the transcription factors PU.1, IRF8, and BATF3, which enabled them to present antigens as type 1 conventional dendritic cells. Reprogrammed tumor cells remodeled their tumor microenvironment, recruited, and expanded polyclonal cytotoxic T cells; induced tumor regressions; and established long-term systemic immunity in multiple mouse melanoma models. In human tumor spheroids and xenografts, reprogramming to immunogenic dendritic-like cells progressed independently of immunosuppression, which usually limits immunotherapy. Our study paves the way for human clinical trials of in vivo immune cell reprogramming for cancer immunotherapy.
Collapse
Affiliation(s)
- Ervin Ascic
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | | | - Malavika Sreekumar Nair
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - André Rosa
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Ilia Kurochkin
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - Olga Zimmermannova
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - Xavier Catena
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | | | | | | | - Tommaso Ballocci
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | | | - Xiaoli Huang
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Maria de Rosa Torres
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
| | - Emilie Renaud
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Marta Velasco Santiago
- National Center of Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730Herlev, Denmark
| | - Özcan Met
- National Center of Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, 2800Kongens Lyngby, Denmark
| | - David Askmyr
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, 221 85Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84Lund, Sweden
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Medicon Village, 223 81Lund, Sweden
| | - Lennart Greiff
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, 221 85Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84Lund, Sweden
| | | | | | - Inge Marie Svane
- National Center of Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730Herlev, Denmark
| | | | - Fábio F. Rosa
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
| | - Carlos-Filipe Pereira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, 221 84Lund, Sweden
- Wallenberg Center for Molecular Medicine at Lund University, 221 84Lund, Sweden
- Asgard Therapeutics AB, Medicon Village, 223 81Lund, Sweden
- Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal, 3004-517Coimbra, Portugal
| |
Collapse
|
276
|
Chhabra Y, Fane ME, Pramod S, Hüser L, Zabransky DJ, Wang V, Dixit A, Zhao R, Kumah E, Brezka ML, Truskowski K, Nandi A, Marino-Bravante GE, Carey AE, Gour N, Maranto DA, Rocha MR, Harper EI, Ruiz J, Lipson EJ, Jaffee EM, Bibee K, Sunshine JC, Ji H, Weeraratna AT. Sex-dependent effects in the aged melanoma tumor microenvironment influence invasion and resistance to targeted therapy. Cell 2024; 187:6016-6034.e25. [PMID: 39243764 PMCID: PMC11580838 DOI: 10.1016/j.cell.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
There is documented sex disparity in cutaneous melanoma incidence and mortality, increasing disproportionately with age and in the male sex. However, the underlying mechanisms remain unclear. While biological sex differences and inherent immune response variability have been assessed in tumor cells, the role of the tumor-surrounding microenvironment, contextually in aging, has been overlooked. Here, we show that skin fibroblasts undergo age-mediated, sex-dependent changes in their proliferation, senescence, ROS levels, and stress response. We find that aged male fibroblasts selectively drive an invasive, therapy-resistant phenotype in melanoma cells and promote metastasis in aged male mice by increasing AXL expression. Intrinsic aging in male fibroblasts mediated by EZH2 decline increases BMP2 secretion, which in turn drives the slower-cycling, highly invasive, and therapy-resistant melanoma cell phenotype, characteristic of the aged male TME. Inhibition of BMP2 activity blocks the emergence of invasive phenotypes and sensitizes melanoma cells to BRAF/MEK inhibition.
Collapse
Affiliation(s)
- Yash Chhabra
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | - Mitchell E Fane
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sneha Pramod
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Laura Hüser
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Daniel J Zabransky
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vania Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Agrani Dixit
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ruzhang Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Edwin Kumah
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Megan L Brezka
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kevin Truskowski
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Asmita Nandi
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gloria E Marino-Bravante
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Naina Gour
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Devon A Maranto
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Murilo R Rocha
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth I Harper
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Justin Ruiz
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Evan J Lipson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA; The Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kristin Bibee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joel C Sunshine
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
277
|
Fujimura T, Yoshino K, Kato H, Fukushima S, Ishizuki S, Otsuka A, Matsushita S, Amagai R, Muto Y, Yamazaki E, Kambayashi Y, Yahata T, Miyata T, Fujisawa Y, Asano Y. A phase II multicentre study of plasminogen activator inhibitor-1 inhibitor (TM5614) plus nivolumab for treating anti-programmed cell death 1 antibody-refractory malignant melanoma: TM5614-MM trial. Br J Dermatol 2024; 191:691-697. [PMID: 38833158 DOI: 10.1093/bjd/ljae231] [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: 04/18/2024] [Revised: 05/12/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Anti-programmed cell death 1 antibodies (PD-1 Abs) are widely used for advanced melanoma, but information on the efficacy of anti-PD-1 Abs is limited in the Asian population. There remains an unmet need to improve the therapeutic effects of anti-PD-1 Ab-treatment, particularly in patients with melanoma who are refractory to anti-PD-1 Abs. The aim of this study was to evaluate anti-PD-1 Ab-treatment in combination with TM5614 (a plasminogen activator inhibitor-1 inhibitor) in patients with unresectable melanoma. METHODS The TM5614-MM study was a multicentre, open-label, single-arm, phase II clinical trial to evaluate the efficacy and safety of nivolumab in combination with TM5614 in patients with advanced, unresectable malignant melanoma recruited at seven Japanese institutes between 13 September 2021 and 31 March 2023. Patients with metastatic or unresectable melanoma previously treated with anti-PD-1 Abs were enrolled. Nivolumab 480 mg was administered intravenously every 4 weeks for 8 weeks, while TM5614 was administered orally at a dose of 120 mg (0-4 weeks) and 180 mg once daily (5-8 weeks). The primary endpoint was the overall response rate after 8 weeks of concomitant use of TM5614. RESULTS Thirty-nine patients were enrolled, and 34 patients were included in the anti-PD-1 Ab-refractory cohort. The overall response rate at 8 weeks was 25.9% (95% confidence interval 12.9-44.9%, P = 0.027) in 27 patients who were anti-PD-1 Ab-refractory based on investigator assessment in the protocol per set cohort. Seven patients discontinued treatment owing to progressive disease or adverse events. Treatment-related grade 3 or higher adverse events occurred in 3 of 39 patients (7.7%) in the intention-to-treat cohort. CONCLUSIONS TM5614 in combination with nivolumab is well tolerated and effective in anti-PD-1 Ab-refractory unresectable melanoma.
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Koji Yoshino
- Department of Dermato-Oncology/Dermatology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo,Japan
- Department of Dermato-Oncology/Dermatology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroshi Kato
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shoichiro Ishizuki
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Otsuka
- Department of Dermatology, Kindai University Hospital, Osaka, Japan
| | - Shigeto Matsushita
- Department of Dermato-Oncology/Dermatology, NHO Kagoshima Medical Center, Kagoshima, Japan
| | - Ryo Amagai
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Yusuke Muto
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Emi Yamazaki
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Takashi Yahata
- Translational Molecular Therapeutic Laboratory, Division of Host Defence Mechanism, Tokai University School of Medicine, Isehara,Japan
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, Tohoku University School of Medicine, Sendai, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Dermatology, Ehime University, Matsuyama, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
278
|
Benguerfi S, Hirsinger B, Raimbourg J, Agbakou M, Muñoz Calahorro R, Vennier A, Lancrey-Javal T, Nedelec P, Seguin A, Reignier J, Lascarrou JB, Canet E. Outcome of patients with solid malignancies considered for intensive care unit admission: a single-center prospective cohort study. Support Care Cancer 2024; 32:726. [PMID: 39397173 DOI: 10.1007/s00520-024-08935-z] [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: 03/21/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE To identify the predictors and outcomes of ICU triage decisions in patients with solid malignancies (SM) and to investigate the usefulness of the National Early Warning Score (NEWS) and quick Sequential Organ Failure Assessment (qSOFA) score at triage. METHODS All patients with SM for whom ICU admission was requested between July 2019 and December 2021 in a French university-affiliated hospital were included prospectively. RESULTS Of the 6262 patients considered for ICU admission, 410 (6.5%) had SM (age, 66 [58-73] years; metastases, 60.1%; and performance status 0-2, 81%). Of these 410 patients, 176 (42.9%) were admitted to the ICU, including 141 (80.1%) subsequently discharged alive. Breast cancer, hemoptysis, and pneumothorax were associated with ICU admission; whereas older age, performance status 3-4, metastatic disease, and request at night were associated with denial of ICU admission. The NEWS, and the qSOFA score in patients with suspected infection, determined at triage performed poorly for predicting hospital mortality (area under the receiver operating characteristics curve, 0.52 and 0.62, respectively). Performance status 3-4 was independently associated with higher 6-month mortality and first-line anticancer treatment with lower 6-month mortality. Hospital mortality was 33.3% in patients admitted to the ICU after refusal of the first request. CONCLUSION Patients with SM were frequently denied ICU admission despite excellent in-ICU survival. Poor performance status was associated with ICU admission denial and higher 6-month mortality, but none of the other reasons for denying ICU admission predicted 6-month mortality. Physiological scores had limited usefulness in this setting.
Collapse
Affiliation(s)
- Soraya Benguerfi
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France.
- ICU, Nantes University, Nantes University Hospital, Movement-Interactions-Performance Research Unit (MIP, UR 4334), Nantes, France.
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire Hôtel-Dieu, 30 Bd. Jean Monnet, 44093, Nantes Cedex 1, France.
| | - Baptiste Hirsinger
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
| | - Judith Raimbourg
- Institut de Cancérologie de L'Ouest, 44805, Saint Herblain, France
| | - Maïté Agbakou
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
| | | | - Alice Vennier
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
| | | | - Paul Nedelec
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
| | - Amélie Seguin
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
| | - Jean Reignier
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
- ICU, Nantes University, Nantes University Hospital, Movement-Interactions-Performance Research Unit (MIP, UR 4334), Nantes, France
| | - Jean-Baptiste Lascarrou
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
- ICU, Nantes University, Nantes University Hospital, Movement-Interactions-Performance Research Unit (MIP, UR 4334), Nantes, France
| | - Emmanuel Canet
- Service de Médecine Intensive Réanimation, CHU de Nantes, Nantes Université, France
| |
Collapse
|
279
|
Huang J, Xiong L, Tang S, Zhao J, Zuo L. Balancing Tumor Immunotherapy and Immune-Related Adverse Events: Unveiling the Key Regulators. Int J Mol Sci 2024; 25:10919. [PMID: 39456702 PMCID: PMC11507008 DOI: 10.3390/ijms252010919] [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: 08/28/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Tumor immunotherapy has emerged as a promising approach in cancer treatment in recent years, offering vast potential. This method primarily involves targeting and inhibiting the suppressive checkpoints present in different immune cells to enhance their activation, ultimately leading to tumor regression. However, tumor cells exploit the surrounding immune cells and tissues to establish a tumor microenvironment (TME) that supports their survival and growth. Within the TME, the efficacy of effector immune cells is compromised, as tumor cells exploit inhibitory immune cells to suppress their function. Furthermore, certain immune cells can be co-opted by tumor cells to facilitate tumor growth. While significantly enhancing the body's tumor immunity can lead to tumor regression, it can also result in severe toxic side effects and an inflammatory factor storm. As a consequence, patients often discontinue treatment due to immune-related adverse events (irAEs) or, in extreme cases, succumb to toxic side effects before experiencing tumor regression. In this analysis, we examined several remission regimens for irAEs, each with its own drawbacks, including toxic side effects or suppression of tumor immunotherapy, which is undesirable. A recent research study, specifically aimed at downregulating intestinal epithelial barrier permeability, has shown promising results in reducing the severity of inflammatory bowel disease (IBD) while preserving immune function. This approach effectively reduces the severity of IBD without compromising the levels of TNF-α and IFN-γ, which are crucial for maintaining the efficacy of tumor immunotherapy. Based on the substantial similarities between IBD and ICI colitis (combo immune checkpoint inhibitors-induced colitis), this review proposes that targeting epithelial cells represents a crucial research direction for mitigating irAEs in the future.
Collapse
Affiliation(s)
- Jianshang Huang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Lei Xiong
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Sainan Tang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Junhao Zhao
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Li Zuo
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| |
Collapse
|
280
|
Picca F, Giannotta C, Tao J, Giordanengo L, Munir HMW, Botta V, Merlini A, Mogavero A, Garbo E, Poletto S, Bironzo P, Doronzo G, Novello S, Taulli R, Bersani F. From Cancer to Immune Organoids: Innovative Preclinical Models to Dissect the Crosstalk between Cancer Cells and the Tumor Microenvironment. Int J Mol Sci 2024; 25:10823. [PMID: 39409152 PMCID: PMC11476904 DOI: 10.3390/ijms251910823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Genomic-oriented oncology has improved tumor classification, treatment options, and patient outcomes. However, genetic heterogeneity, tumor cell plasticity, and the ability of cancer cells to hijack the tumor microenvironment (TME) represent a major roadblock for cancer eradication. Recent biotechnological advances in organotypic cell cultures have revolutionized biomedical research, opening new avenues to explore the use of cancer organoids in functional precision oncology, especially when genomics alone is not a determinant. Here, we outline the potential and the limitations of tumor organoids in preclinical and translational studies with a particular focus on lung cancer pathogenesis, highlighting their relevance in predicting therapy response, evaluating treatment toxicity, and designing novel anticancer strategies. Furthermore, we describe innovative organotypic coculture systems to dissect the crosstalk with the TME and to test the efficacy of different immunotherapy approaches, including adoptive cell therapy. Finally, we discuss the potential clinical relevance of microfluidic mini-organ technology, capable of reproducing tumor vasculature and the dynamics of tumor initiation and progression, as well as immunomodulatory interactions among tumor organoids, cancer-associated fibroblasts (CAFs) and immune cells, paving the way for next-generation immune precision oncology.
Collapse
Affiliation(s)
- Francesca Picca
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
- Molecular Biotechnology Center ‘Guido Tarone’, University of Torino, Piazza Nizza 44, 10126 Torino, Italy
| | - Claudia Giannotta
- Molecular Biotechnology Center ‘Guido Tarone’, University of Torino, Piazza Nizza 44, 10126 Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Jiahao Tao
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Lucia Giordanengo
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - H. M. Waqas Munir
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Virginia Botta
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Alessandra Merlini
- Thoracic Unit and Medical Oncology Division, Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Andrea Mogavero
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Edoardo Garbo
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Stefano Poletto
- Thoracic Unit and Medical Oncology Division, Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Paolo Bironzo
- Thoracic Unit and Medical Oncology Division, Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Silvia Novello
- Thoracic Unit and Medical Oncology Division, Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
| | - Riccardo Taulli
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
- Molecular Biotechnology Center ‘Guido Tarone’, University of Torino, Piazza Nizza 44, 10126 Torino, Italy
| | - Francesca Bersani
- Department of Oncology, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole 10, 10043 Orbassano, Italy
- Molecular Biotechnology Center ‘Guido Tarone’, University of Torino, Piazza Nizza 44, 10126 Torino, Italy
| |
Collapse
|
281
|
Gradone AL, Ma VT, Vasbinder A, Fecher LA, Yentz S, Hayek SS, Lao CD. Increased myositis and possible myocarditis in melanoma patients treated with immune checkpoint inhibitors in the COVID-19 era. Cancer Immunol Immunother 2024; 73:259. [PMID: 39369180 PMCID: PMC11456101 DOI: 10.1007/s00262-024-03803-5] [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: 12/07/2023] [Accepted: 08/08/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI)-mediated myocarditis results in significant morbidity and mortality. At our institution, we noted an increased incidence of ICI-mediated myocarditis cases, leading to further investigation in our database of advanced melanoma patients treated with ICI therapy. METHODS A single-center, retrospective cohort analysis of patients with advanced melanoma identified cases of ICI-mediated myocarditis and myositis. RESULTS 366 patients with advanced melanoma received a dose of ICI from September 2014 to October 2019. Of these patients, there were 0 cases of ICI-mediated myocarditis (0%, 95% CI 0%-1.0%) and 2 cases of ICI-mediated myositis (0.55%, 95% CI 0.07%-1.96%). From November 2019 to December 2021, an additional 246 patients with advanced melanoma were identified. Of these patients, 10 (4.1%, 95% CI 1.97%-7.35%) developed ICI-mediated myocarditis and 10 developed ICI-mediated myositis. CONCLUSION Our study suggests an increase in prevalence of ICI-mediated muscle damage including myositis and myocarditis in the COVID-19 era. Differentiation of these patients and further risk stratification may allow for development of guidelines for nuanced management of this serious complication.
Collapse
Affiliation(s)
- Allison L Gradone
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Vincent T Ma
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Internal Medicine, University of Wisconsin, Madison, WI, USA
- Department of Dermatology, University of Wisconsin, Madison, WI, USA
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leslie A Fecher
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Yentz
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christopher D Lao
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
282
|
Tang B, Chen Y, Jiang Y, Fang M, Gao Q, Ren X, Yao L, Huang G, Chen J, Zhang X, Li R, Zhao S, Gao M, Luo R, Qi M, Li F, Zheng F, Lee M, Tao X, Duan R, Guo J, Chi Z, Cui C. Toripalimab in combination with HBM4003, an anti-CTLA-4 heavy chain-only antibody, in advanced melanoma and other solid tumors: an open-label phase I trial. J Immunother Cancer 2024; 12:e009662. [PMID: 39366752 PMCID: PMC11459314 DOI: 10.1136/jitc-2024-009662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND HBM4003 is a novel anti-CTLA-4 heavy chain-only antibody, designed to enhance Treg ablation and antibody-dependent cell-mediated cytotoxicity while ensuring a manageable safety profile. This phase I trial investigated the safety, pharmacokinetics, immunogenicity and preliminary efficacy of HBM4003 plus with anti-PD-1 antibody toripalimab in patients with advanced solid tumors, especially focusing on melanoma. METHODS The multicenter, open-label phase I trial was divided into two parts: dose-escalation phase (part 1) and dose-expansion phase (part 2). In part 1, HBM4003 was administered at doses of 0.03, 0.1, 0.3 mg/kg in combination with toripalimab with fixed dosage of 240 mg every 3 weeks. The recommended phase II dose (RP2D) was used in the expansion phase. Primary endpoints were safety and RP2D in part 1 and objective response rate (ORR) in part 2. Biomarkers based on cytokines and multiplex immunofluorescence staining were explored. RESULTS A total of 40 patients received study treatment, including 36 patients treated with RP2D of HBM4003 0.3 mg/kg plus toripalimab 240 mg every 3 week. 36 participants (90.0%) experienced at least one treatment-related adverse event (TRAE), of which 10 (25.0%) patients experienced grade ≥3 TRAEs and 5 (12.5%) experienced immune-mediated adverse events (irAEs) with maximum severity of grade 3. No grade 4 or 5 irAEs occurred. Efficacy analysis set included 32 melanoma patients treated with RP2D and with available post-baseline imaging data. The ORRs of anti-PD-1/PD-L1 treatment-naïve subgroup and anti-PD-1/PD-L1 treatment-failed subgroup were 33.3% and 5.9%, respectively. In mucosal melanoma, the ORR of the two subgroups were 40.0% and 10.0%, respectively. Baseline high Treg/CD4+ratio in the tumor serves as an independent predictive factor for the efficacy of immunotherapy. CONCLUSIONS HBM4003 0.3 mg/kg plus toripalimab 240 mg every 3 week demonstrated manageable safety in solid tumors and no new safety signal. Limited data demonstrated promising antitumor activity, especially in PD-1 treatment-naïve mucosal melanoma. TRIAL REGISTRATION NUMBER NCT04727164.
Collapse
Affiliation(s)
- Bixia Tang
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Yu Jiang
- Department of Head and Neck Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Meiyu Fang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Quanli Gao
- Immunotherapy Department, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Yao
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Gang Huang
- Central South University (Hunan Cancer Hospital), Changsha, Hunan, China
| | - Jing Chen
- Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoshi Zhang
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rongqing Li
- Tumor Radiotherapy Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | | | | | | | - Meng Qi
- Harbour BioMed, Shanghai, China
| | - Feng Li
- Harbour BioMed, Shanghai, China
| | | | | | | | - Rong Duan
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
283
|
Verkhovskaia S, Falcone R, Di Pietro FR, Carbone ML, Samela T, Perez M, Poti G, Morelli MF, Zappalà AR, Di Rocco ZC, Morese R, Piesco G, Chesi P, Marchetti P, Abeni D, Failla CM, De Galitiis F. Survival of Patients with Metastatic Melanoma Treated with Ipilimumab after PD-1 Inhibitors: A Single-Center Real-World Study. Cancers (Basel) 2024; 16:3397. [PMID: 39410017 PMCID: PMC11475497 DOI: 10.3390/cancers16193397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND When monotherapy with PD-1 inhibitors in metastatic melanoma fails, there are currently no standard second-line choices. In case of the unavailability of clinical trials, ipilimumab represents a possible alternative treatment. METHODS We collected data of 44 patients who received ipilimumab after the failure of PD-1 inhibitors from July 2017 to May 2023 at our Institute. Overall survival (OS), progression-free survival (PFS), and post-progression survival (PPS) based on BRAF or NRAS mutation status, sex, and the presence of brain metastases were estimated using the Kaplan-Meier method. Cox regression was used to evaluate independence in multivariate analysis. The objective response rate (ORR) was estimated based on RECIST 1.1. RESULTS Among the 44 patients enrolled in this study, 28 BRAF-wildtype, 9 BRAF-mutated, and 7 NRAS-mutated patients were identified. OS analysis showed a significant difference between wildtype and BRAF- or NRAS-mutated patients: 23.2 months vs 5.3 and 4.59, respectively, p = 0.017. The presence of brain metastases and BRAF or NRAS mutation were independent factors for mortality in multivariate analysis. CONCLUSIONS In case of failure to enroll patients in innovative clinical trials, second-line ipilimumab still represents an effective therapy in patients with metastatic wildtype melanoma and in the absence of brain metastases.
Collapse
Affiliation(s)
- Sofia Verkhovskaia
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Rosa Falcone
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Francesca Romana Di Pietro
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Maria Luigia Carbone
- Clinical Trial Center, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy
| | - Tonia Samela
- Epidemiology Units, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (T.S.); (D.A.)
| | - Marie Perez
- Department of Histopathology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy;
| | - Giulia Poti
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Maria Francesca Morelli
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Albina Rita Zappalà
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Zorika Christiana Di Rocco
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Roberto Morese
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Gabriele Piesco
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Paolo Chesi
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Paolo Marchetti
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Damiano Abeni
- Epidemiology Units, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (T.S.); (D.A.)
| | - Cristina Maria Failla
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy;
| | - Federica De Galitiis
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| |
Collapse
|
284
|
Shaie KM, Sihan L, Yuli W, Mengfei H, Renqian F, Yan H. Knowledge mapping and visualization of trends in immunotherapy for ovarian cancer over the past five years: a bibliometric analysis. Front Immunol 2024; 15:1465917. [PMID: 39421747 PMCID: PMC11483997 DOI: 10.3389/fimmu.2024.1465917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background This study conducts a bibliometric literature analysis to explore trends in immunotherapy for ovarian cancer from 2019 to 2023. Methods An extensive online literature search was conducted in the Web of Science Core Collection database to identify English-language articles and reviews related to "trends in immunotherapy", and "ovarian cancer". statistical analysis was performed using VOSviewer to visualize and compare nations, institutions, and journals simultaneously. Results Our findings highlight contributions by 118 nations, led by the People's Republic of China with 3,167 contributions; Germany followed with 558 and Italy having 547. Of all publications made between 2019-2023, "Frontiers Immunology" had the most publications with 546 total records followed by "cancers ", and "frontiers in oncology" being the most heavily relied upon categories. Annually publication trends increased until 2022 but then declined considerably as a peak of highly-cited papers occurring between 2019 and 2022. Conclusions Our bibliometric analysis not only maps the evolution of immunotherapy research in ovarian cancer but also provides actionable insights for advancing scientific progress. By identifying emerging trends and key areas, future research can strategically enhance treatment strategies and outcomes for ovarian cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Hu Yan
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
285
|
Fokken C, Silbern I, Shomroni O, Pan KT, Ryazanov S, Leonov A, Winkler N, Urlaub H, Griesinger C, Becker D. Interfering with aggregated α-synuclein in advanced melanoma leads to a major upregulation of MHC class II proteins. Melanoma Res 2024; 34:393-407. [PMID: 38950202 PMCID: PMC11361348 DOI: 10.1097/cmr.0000000000000982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/03/2024] [Indexed: 07/03/2024]
Abstract
Melanoma is the most serious and deadly form of skin cancer and with progression to advanced melanoma, the intrinsically disordered protein α-synuclein is upregulated to high levels. While toxic to dopaminergic neurons in Parkinson's disease, α-synuclein is highly beneficial for primary and metastatic melanoma cells. To gain detailed insights into this exact opposite role of α-synuclein in advanced melanoma, we performed proteomic studies of high-level α-synuclein-expressing human melanoma cell lines that were treated with the diphenyl-pyrazole small-molecule compound anle138b, which binds to and interferes with the oligomeric structure of α-synuclein. We also performed proteomic and transcriptomic studies of human melanoma xenografts that were treated systemically with the anle138b compound. The results reveal that interfering with oligomerized α-synuclein in the melanoma cells in these tumor xenografts led to a substantial upregulation and expression of major histocompatibility complex proteins, which are pertinent to enhancing anti-melanoma immune responses.
Collapse
Affiliation(s)
- Claudia Fokken
- Department of NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences
- Bioanalytics Research Group, Institute of Clinical Chemistry, University Medical Center Göttingen
| | - Orr Shomroni
- NGS-Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Center Göttingen
| | - Kuan-Ting Pan
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences
| | - Sergey Ryazanov
- Department of NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences
| | - Andrei Leonov
- Department of NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences
| | - Nadine Winkler
- Department of NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences
- Bioanalytics Research Group, Institute of Clinical Chemistry, University Medical Center Göttingen
| | - Christian Griesinger
- Department of NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), Georg-August-University Göttingen
| | - Dorothea Becker
- Department of NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences
- Institute for Organic and Biomolecular Chemistry, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
286
|
Therien AD, Chime-Eze CM, Rhodin KE, Beasley GM. Neoadjuvant therapy for melanoma: past, present, and future. Surg Oncol 2024; 56:102127. [PMID: 39236515 DOI: 10.1016/j.suronc.2024.102127] [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: 06/07/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
Modern systemic therapy has dramatically improved outcomes for many patients with advanced metastatic melanoma. The success of these therapies has attracted much scientific interest while these therapies have made their way into the treatment of earlier stages of disease. Randomized trials have led to the approval of adjuvant immunotherapy and targeted therapy for resected stage III melanoma. However, most recently, these therapies have gained traction in the neoadjuvant setting. Promising early results led to randomized controlled trials that have now established neoadjuvant therapy as standard of care in advanced melanoma patients. Questions remain regarding the optimal choice of therapy, duration and timing of neoadjuvant therapy, extent of surgery, and the need for additional adjuvant therapy for patients who received neoadjuvant therapy. Herein we provide an overview of neoadjuvant therapy for melanoma and dilemmas to its broader applications.
Collapse
Affiliation(s)
| | | | - Kristen E Rhodin
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
287
|
Lodde GC, Zhao F, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Jansen P, Kowall B, Galetzka W, Hörst F, Kleesiek J, Hellwig B, Rahnenführer J, Rajcsanyi L, Peters T, Hinney A, Placke JM, Sucker A, Paschen A, Becker JC, Livingstone E, Zimmer L, Tasdogan A, Roesch A, Hadaschik E, Schadendorf D, Griewank K, Ugurel S. Early versus late response to PD-1-based immunotherapy in metastatic melanoma. Eur J Cancer 2024; 210:114295. [PMID: 39213786 DOI: 10.1016/j.ejca.2024.114295] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Immune checkpoint inhibition (ICI) currently is the most effective treatment to induce durable responses in metastatic melanoma. The aims of this study are the characterization of patients with early, late and non-response to ICI and analysis of survival outcomes in a real-world patient cohort. METHODS Patients who received PD-1-based immunotherapy for non-resectable stage-IV melanoma in any therapy line were selected from the prospective multicenter real-world DeCOG study ADOREG-TRIM (NCT05750511). Patients showing complete (CR) or partial (PR) response already during the first 3 months of treatment (Early Responders, EarlyR) were compared to patients showing CR/PR at a later time (Late Responders, LateR), a stable disease (SD) and to patients showing progressive disease (Non-Responders, NonR). RESULTS Of 522 patients, 8.2 % were EarlyR (n = 43), 19.0 % were LateR (n = 99), 37.0 % had a SD (n = 193) and 35.8 % were NonR (n = 187). EarlyR, LateR and SD patients had comparable baseline characteristics. Multivariate logbinomial regression analyses adjusted for age and sex revealed positive tumor PD-L1 (RR=1.99, 95 %-CI=1.14-3.46, p = 0.015), and normal serum CRP (RR=1.59, 95 %-CI=0.93-2.70, p = 0.036) as independently associated with the achievement of an early response compared to NonR. The median progression-free and overall survival was 46.0 months (95 % CI 19.1; NR) and 47.8 months (95 %-CI 36.9; NR) for EarlyR, NR (95 %-CI NR; NR) for LateR, 8.1 months (7.0; 10.4) and 35.4 months (29.2; NR) for SD, and 2.0 months (95 %-CI 1.9; 2.1) and 6.1 months (95 %-CI 4.6; 8.8) for NonR patients. CONCLUSION Less than 10 % of metastatic melanoma patients achieved an early response during the first 3 months of PD-1-based immunotherapy. Early responders were not superior to late responders in terms of response durability and survival.
Collapse
Affiliation(s)
- Georg C Lodde
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Fang Zhao
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Rudolf Herbst
- Department of Dermatology, Helios Klinikum Erfurt, Erfurt, Germany.
| | | | - Jochen Utikal
- Department of Dermatology, Venerology, and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, 68167 Mannheim, Germany.
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg, Saar, Germany.
| | - Jens Ulrich
- Department of Dermatology and Venereology, Harzklinikum Dorothea Christiane Erxleben, Quedlinburg, Germany.
| | - Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Klinik Oberhausen, University Witten/Herdecke, Oberhausen, Germany.
| | - Peter Mohr
- Dermatological Center Buxtehude, Elbe Kliniken Buxtehude, Buxtehude, Germany.
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center Minden, Ruhr University Bochum, Minden, Germany.
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, Dresden, Germany.
| | - Edgar Dippel
- Department of Dermatology Ludwigshafen, Klinikum der Stadt Ludwigshafen am Rhein gGmbH, Ludwigshafen, Germany.
| | - Michael Weichenthal
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany.
| | - Philipp Jansen
- Department of Dermatology, University Hospital Bonn, Bonn, Germany.
| | - Bernd Kowall
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany.
| | - Wolfgang Galetzka
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany.
| | - Fabian Hörst
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany.
| | - Jens Kleesiek
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany.
| | - Birte Hellwig
- Department of Statistics, TU Dortmund University, Dortmund, Germany.
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, Dortmund, Germany.
| | - Luisa Rajcsanyi
- Section for Molecular Genetics of Mental Disorders, University Hospital Essen, Essen Essen, Germany Center for Translational Neuro, and Behavioral Sciences, University Hospital Essen, Essen, Germany.
| | - Triinu Peters
- Section for Molecular Genetics of Mental Disorders, University Hospital Essen, Essen Essen, Germany Center for Translational Neuro, and Behavioral Sciences, University Hospital Essen, Essen, Germany.
| | - Anke Hinney
- Section for Molecular Genetics of Mental Disorders, University Hospital Essen, Essen Essen, Germany Center for Translational Neuro, and Behavioral Sciences, University Hospital Essen, Essen, Germany.
| | - Jan-Malte Placke
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| | - Antje Sucker
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Annette Paschen
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| | - Jürgen C Becker
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany; Translational Skin Cancer Research, German Consortium for Translational Cancer Research (DKTK), Essen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Lisa Zimmer
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| | - Alpaslan Tasdogan
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| | - Alexander Roesch
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| | - Eva Hadaschik
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| | - Klaus Griewank
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), Essen, Düsseldorf, Germany.
| |
Collapse
|
288
|
Huang W, Kim BS, Zhang Y, Lin L, Chai G, Zhao Z. Regulatory T cells subgroups in the tumor microenvironment cannot be overlooked: Their involvement in prognosis and treatment strategy in melanoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:4512-4530. [PMID: 38530049 DOI: 10.1002/tox.24247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Melanoma, the most lethal form of skin cancer, presents substantial challenges despite effective surgical interventions for in situ lesions. Regulatory T cells (Tregs) wield a pivotal immunomodulatory influence within the tumor microenvironment, yet their impact on melanoma prognosis and direct molecular interactions with melanoma cells remain elusive. This investigation employs single-cell analysis to unveil the intricate nature of Tregs in human melanoma. METHODS Single-cell RNA and bulk sequencing data, alongside clinical information, were obtained from public repositories. Initially, GO and GSEA analyses were employed to delineate functional disparities among distinct cell subsets. Pseudotime and cell-cell interconnection analyses were conducted, followed by an endeavor to construct a prognostic model grounded in Treg-associated risk scores. This model's efficacy was demonstrated via PCA and K-M analyses, with multivariate Cox regression affirming its independent prognostic value in melanoma patients. Furthermore, immune infiltration analysis, immune checkpoint gene expression scrutiny, and drug sensitivity assessments were performed to ascertain the clinical relevance of this prognostic model. RESULTS Following batch effect correction, 80 025 cells partitioned into 31 clusters, encompassing B cells, plasma cells, endothelial cells, fibroblasts, melanoma cells, monocytes, macrophages, and T_NK cells. Within these, 4240 CD4+ T cells were subclassified into seven distinct types. Functional analysis underscored the immunomodulatory function of Tregs within the melanoma tumor microenvironment, elucidating disparities among Treg subpopulations. Notably, the ITGB2 signaling pathway emerged as a plausible molecular nexus linking Tregs to melanoma cells. Our prognostic signature exhibited robust predictive capacities for melanoma prognosis and potential implications in evaluating immunotherapy response. CONCLUSION Tregs exert a critical role in immune suppression within the melanoma tumor microenvironment, revealing a potential molecular-level association with melanoma cells. Our innovative Treg-centered signature introduces a promising prognostic marker for melanoma, holding potential for future clinical prognostic assessments.
Collapse
Affiliation(s)
- Wenyi Huang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Byeong Seop Kim
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Lin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Stomatology, First Affiliated Hospital of Soochow University, Suzhou, China
- National Center for Translational Medicine(Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Gang Chai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
289
|
Park SJ, Kweon S, Moyo MK, Kim HR, Choi JU, Lee NK, Maharjan R, Cho YS, Park JW, Byun Y. Immune modulation of the liver metastatic colorectal cancer microenvironment via the oral CAPOX-mediated cGAS-STING pathway. Biomaterials 2024; 310:122625. [PMID: 38820768 DOI: 10.1016/j.biomaterials.2024.122625] [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: 01/30/2024] [Revised: 05/05/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
We evaluated modulation of the immunosuppressive tumor microenvironment in both local and liver metastatic colorectal cancer (LMCC), focusing on tumor-associated macrophages, which are the predominant immunosuppressive cells in LMCC. We developed an orally administered metronomic chemotherapy regimen, oral CAPOX. This regimen combines capecitabine and a nano-micelle encapsulated, lysine-linked deoxycholate and oxaliplatin complex (OPt/LDC-NM). The treatment effectively modulated immune cells within the tumor microenvironment by activating the cGAS-STING pathway and inducing immunogenic cell death. This therapy modulated immune cells more effectively than did capecitabine monotherapy, the current standard maintenance chemotherapy for colorectal cancer. The macrophage-modifying effect of oral CAPOX was mediated via the cGAS-STING pathway. This is a newly identified mode of immune cell activation induced by metronomic chemotherapy. Moreover, oral CAPOX synergized with anti-PD-1 antibody (αPD-1) to enhance the T-cell-mediated antitumor immune response. In the CT26. CL25 subcutaneous model, combination therapy achieved a 91 % complete response rate with a confirmed memory effect against the tumor. This combination also altered the immunosuppressive tumor microenvironment in LMCC, which αPD-1 monotherapy could not achieve. Oral CAPOX and αPD-1 combination therapy outperformed the maximum tolerated dose for treating LMCC, suggesting metronomic therapy as a promising strategy.
Collapse
Affiliation(s)
- Seong Jin Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | | | - Ha Rin Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; School of Medicine, Oncology, Stanford University, CA, 94305, United States
| | - Jeong Uk Choi
- College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Na Kyeong Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Ruby Maharjan
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, United States
| | - Young Seok Cho
- College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Jin Woo Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
290
|
Ascierto PA, Mandalà M, Ferrucci PF, Guidoboni M, Rutkowski P, Ferraresi V, Arance A, Guida M, Maiello E, Gogas H, Richtig E, Quaglino P, Lebbé C, Helgadottir H, Queirolo P, Spagnolo F, Tucci M, Del Vecchio M, Gonzalez-Cao M, Minisini AM, De Placido S, Sanmamed MF, Casula M, Bulgarelli J, Pisano M, Piccinini C, Piccin L, Cossu A, Mallardo D, Paone M, Vitale MG, Melero I, Grimaldi AM, Giannarelli D, Palmieri G, Dummer R, Sileni VC. Sequencing of Checkpoint or BRAF/MEK Inhibitors on Brain Metastases in Melanoma. NEJM EVIDENCE 2024; 3:EVIDoa2400087. [PMID: 39315864 DOI: 10.1056/evidoa2400087] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
BACKGROUND The impact of the order of treatment with checkpoint inhibitors or BRAF/MEK inhibitors on the development of brain metastases in patients with metastatic unresectable BRAFV600-mutant melanoma is unknown. The SECOMBIT trial examined the impact of the order of receipt of these treatments in such patients. METHODS In this three-arm trial, we reviewed patients without brain metastases who received the BRAF/MEK inhibitors encorafenib and binimetinib until they had progressive disease followed by the immune checkpoint inhibitors ipilimumab and nivolumab (arm A); or treatment with ipilimumab and nivolumab until they had progressive disease followed by encorafenib and binimetinib (arm B); or treatment with encorafenib and binimetinib for 8 weeks followed by ipilimumab and nivolumab until they had progressive disease followed by retreatment with encorafenib arm binimetinib (arm C). RESULTS Brain metastases were discovered during the trial in 23/69 patients in arm A, 11/69 in arm B, and 9/68 in arm C. At a median follow-up of 56 months, the 60-month brain metastases-free survival rates were 56% for arm A, 80% for arm B (hazard ratio [HR] vs. A: 0.40, 95% confidence interval [CI] 0.23 to 0.58), and 85% for arm C (HR vs. A: 0.35, 95% CI 0.16 to 0.76). CONCLUSIONS In patients with unresectable metastatic melanoma, the treatment sequence of immune checkpoint inhibition followed by BRAF/MEK inhibitors was associated with longer periods of new brain metastases-free survival than the reverse sequence. A regimen in which immune checkpoint inhibition was sandwiched between BRAF/MEK inhibition also appeared to be protective against brain metastases. (ClinicalTrials.gov number NCT02631447.).
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Mario Mandalà
- Department of Oncologic, Hematologic, Gastroenterological Sciences, University of Perugia, Perugia, Italy
- Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | | | - Massimo Guidoboni
- Experimental and Clinical Oncology of Immunotherapy and Rare Cancers Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori," Meldola, Italy
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie National Research Institute of Oncology, 02-781, Warsaw, Poland
| | - Virginia Ferraresi
- Sarcomas and Rare Tumors Departmental Unit, IRCCS Regina Elena National Cancer Institute, Rome
| | - Ana Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona
| | - Michele Guida
- Rare Tumors and Melanoma Unit, IRCCS Istituto dei Tumori "Giovanni Paolo II," Bari, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens
| | - Erika Richtig
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Pietro Quaglino
- Department of Medical Sciences, Dermatologic Clinic, University of Turin, Turin, Italy
| | - Céleste Lebbé
- Université Paris Cite, Dermato-Oncology and CIC AP-HP Hôpital Saint Louis, Cancer Institute APHP, Nord-Université Paris Cite, INSERM U976, Paris
| | - Hildur Helgadottir
- Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm
| | - Paola Queirolo
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Division of Melanoma, Sarcomas and Rare Tumors, IRCCS European Institute of Oncology, Milan
| | - Francesco Spagnolo
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genova, Italy
| | - Marco Tucci
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro," Bari, Italy
| | - Michele Del Vecchio
- Unit of Melanoma Medical Oncology, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Maria Gonzalez-Cao
- Department of Medical Oncology, Oncology Institute Rosell, University Hospital Dexeus, Barcelona
| | - Alessandro Marco Minisini
- Academic Hospital "Santa Maria della Misericordia," Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," Naples, Italy
| | - Miguel F Sanmamed
- Department of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Milena Casula
- Immuno-Oncology & Targeted Cancer Biotherapies, Unit of Cancer Genetics, University of Sassari, IRGB-CNR, 07100 Sassari, Italy
| | - Jenny Bulgarelli
- Experimental and Clinical Oncology of Immunotherapy and Rare Cancers Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori," Meldola, Italy
| | - Marina Pisano
- Immuno-Oncology & Targeted Cancer Biotherapies, Unit of Cancer Genetics, University of Sassari, IRGB-CNR, 07100 Sassari, Italy
| | - Claudia Piccinini
- Experimental and Clinical Oncology of Immunotherapy and Rare Cancers Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori," Meldola, Italy
| | - Luisa Piccin
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Antonio Cossu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Domenico Mallardo
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Miriam Paone
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Maria Grazia Vitale
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Ignacio Melero
- Department of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Antonio M Grimaldi
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
- Medical Oncology Unit, AORN San Pio, Benevento, Italy
| | - Diana Giannarelli
- Facility of Epidemiology and Biostatistics, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome
| | - Giuseppe Palmieri
- Immuno-Oncology & Targeted Cancer Biotherapies, Unit of Cancer Genetics, University of Sassari, IRGB-CNR, 07100 Sassari, Italy
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
291
|
Maurer A, Clerici G, Schaab JA, Cheng PF, Mihic-Probst D, Mader C, Messerli M, Huellner MW, Dummer R, Dimitriou F. Immunotherapy response and resistance in patients with advanced uveal melanoma: a retrospective cohort study. Clin Exp Med 2024; 24:234. [PMID: 39352553 PMCID: PMC11445343 DOI: 10.1007/s10238-024-01497-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Metastatic uveal melanoma (mUM) is associated with poor prognosis. Ipilimumab/nivolumab has shown antitumor efficacy in phase II studies. Tebentafusp resulted in longer overall survival (OS) compared to investigator`s choice in a phase III study. We sought to describe the radiological response patterns of mUM patients treated with immunotherapy. Patients with mUM treated with ipilimumab/nivolumab and tebentafusp between July 2018 and December 2022, with available radiological assessment per RECISTv1.1 and/or imPERCIST5, were retrospectively identified and included. Progression-free survival (PFS) and OS rates, liver-specific response and pathological assessment in available liver biopsies were evaluated. In the ipilimumab/nivolumab group, median PFS (mPFS) was 2.9 months (95% CI 2.2-28.6) and mOS 28.9 months (95% CI 12.7-NR). Complete (CMR) and partial (PMR) metabolic response per imPERCIST5, and partial response (PR) per RECISTv1.1 were associated with longer PFS and OS by trend, compared to morphologically and metabolically stable or progressive disease. In the tebentafusp group, mPFS was 2.7 months (95% CI 2.2-3) and mOS 18.6 months (95% CI 11.5-NR). PMR and PR were associated with longer PFS by trend. In both treatments, the overall treatment response was associated with the radiological response at the liver site. In available liver tumor biopsies, differences in pathological and radiological responses were noted. ImPERCIST5 and RECIST v1.1 are valuable tools in the radiological response assessment, but both methods display limitations. Accurate biomarkers to stratify patients at risk for disease progression and future translational studies to investigate mechanisms of response and resistance are required.
Collapse
Affiliation(s)
- Alexander Maurer
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Giulio Clerici
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Jan A Schaab
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Phil F Cheng
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Daniela Mihic-Probst
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cäcilia Mader
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Michael Messerli
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
292
|
Shen S, Hong Y, Huang J, Qu X, Sooranna SR, Lu S, Li T, Niu B. Targeting PD-1/PD-L1 in tumor immunotherapy: Mechanisms and interactions with host growth regulatory pathways. Cytokine Growth Factor Rev 2024; 79:16-28. [PMID: 39179486 DOI: 10.1016/j.cytogfr.2024.08.001] [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: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Tumor immunotherapy has garnered considerable attention, emerging as a new standard of care in cancer treatment. The conventional targets, such as VEGF and EGFR, have been extended to others including BRAF and PD-1/PD-L1, which have shown significant potential in recent cancer treatments. This review aims to succinctly overview the impact and mechanisms of therapies that modulate PD-1/PD-L1 expression by targeting VEGF, EGFR, LAG-3, CTLA-4 and BRAF. We investigated how modulation of PD-1/PD-L1 expression impacts growth factor signaling, shedding light on the interplay between immunomodulatory pathways and growth factor networks within the tumor microenvironment. By elucidating these interactions, we aim to provide insights into novel potential synergistic therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Songyu Shen
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Yihan Hong
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Jiajun Huang
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Xiaosheng Qu
- Guangxi Botanical Garden of Medicinal Plants, Nanning, Guangxi 530023, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Imperial College London, 369 Fulham Road, London SW10 9NH, United Kingdom
| | - Sheng Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 169 Changle West Rd, Xi'an 710032, China.
| | - Bing Niu
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China.
| |
Collapse
|
293
|
Carey AE, Weeraratna AT. Entering the TiME machine: How age-related changes in the tumor immune microenvironment impact melanoma progression and therapy response. Pharmacol Ther 2024; 262:108698. [PMID: 39098769 DOI: 10.1016/j.pharmthera.2024.108698] [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: 02/29/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
Melanoma is the deadliest form of skin cancer in the United States, with its incidence rates rising in older populations. As the immune system undergoes age-related changes, these alterations can significantly influence tumor progression and the effectiveness of cancer treatments. Recent advancements in understanding immune checkpoint molecules have paved the way for the development of innovative immunotherapies targeting solid tumors. However, the aging tumor microenvironment can play a crucial role in modulating the response to these immunotherapeutic approaches. This review seeks to examine the intricate relationship between age-related changes in the immune system and their impact on the efficacy of immunotherapies, particularly in the context of melanoma. By exploring this complex interplay, we hope to elucidate potential strategies to optimize treatment outcomes for older patients with melanoma, and draw parallels to other cancers.
Collapse
Affiliation(s)
- Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
294
|
Gurel Z, Luy MS, Luo Q, Arp NL, Erbe AK, Kesarwala AH, Fan J, Kimple RJ. Metabolic modulation of melanoma enhances the therapeutic potential of immune checkpoint inhibitors. Front Oncol 2024; 14:1428802. [PMID: 39435293 PMCID: PMC11491500 DOI: 10.3389/fonc.2024.1428802] [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: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Lactate is a pivotal molecule with diverse functions in the metabolic reprogramming of cancer cells. Beyond its role in metabolism, lactate exerts a modulatory effect within the tumor microenvironment; it is utilized by stromal cells and has been implicated in the suppression of the immune response against the tumor. Methods Using in vitro assays (including flow cytometry, live-cell imaging and metabolic analyses), the impact of lactate dehydrogenase inhibitors (LDHIs) on melanoma cells were assessed. The therapeutic potential of LDHIs with immune checkpoint inhibitors (ICIs) were tested in vivo in murine models of melanoma tumors. Results A potent anti-proliferative effect (via both cell cycle alterations and enhanced apoptosis) of LDHIs, Oxamate (Oxa) and methyl 1-hydroxy-6-phenyl-4-(trifluoromethyl)-1H-indole-2-carboxylate (NHI-2), was found upon treatment of melanoma cell lines. Using a combination of Oxa and NHI-2, a synergistic effect to inhibit proliferation, glycolysis, and ATP production was observed. Metabolic analysis revealed significant alteration in glycolysis and oxidative phosphorylation, while metabolite profiling emphasized consequential effects on lactate metabolism and induced energy depletion by LDHIs. Detection of increased RANTES and MCP-1, with Oxa and NHI-2 treatment, prompted the consideration of combining LDHIs with ICIs. In vivo studies using a murine B78 melanoma tumor model revealed a significant improvement in treatment efficacy when LDHIs were combined with ICIs. Conclusions These findings propose the potential of targeting lactate metabolism to enhance the efficacy of ICI treatments in patients with melanoma.
Collapse
Affiliation(s)
- Zafer Gurel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Michael S. Luy
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Qianyun Luo
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Nicholas L. Arp
- Morgridge Institute for Research, Madison, WI, United States
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Aparna H. Kesarwala
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Randall J. Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- University of Wisconsin (UW) Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
295
|
Dini F, Susini P, Zuccaro B, Nisi G, Cuomo R, Grimaldi L, Perillo G, Tinunin L, Antonini P, Innocenti A, Cecchi G, Gambale E, Doni L, Mazzini C, Santoro N, De Giorgi V. Head and neck melanoma: the eyelid region has a better prognosis and easier management. A retrospective survey and systematic review. Melanoma Res 2024; 34:429-438. [PMID: 38833343 DOI: 10.1097/cmr.0000000000000984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Eyelid melanoma (EM) is a malignant neoplasm accounting for around 1% of eyelid malignancies. Because of its rarity, most of our knowledge of EM is currently based on studies of cutaneous melanomas located elsewhere. Accordingly, this study aimed to specifically evaluate EM characteristics, management strategies, and prognosis. A retrospective study was carried out on patients diagnosed with EM at Careggi University Hospital, Florence between May 2012 and May 2022. In addition, a systematic review of relevant literature was conducted, encompassing studies published from 2013 to 2023. Clinical, histopathological, therapeutical, and prognostic data were analyzed to assess the metastasis rate and the 5-year survival rate of patients with EM. Separate data were extracted for in situ and invasive disease. Our original study included 19 patients diagnosed with EM with a 5-year survival rate of 100% for in situ and 83.3% for invasive EM. The literature review identified five poorly detailed large database reviews and 14 original studies on EM with an overall 5-year survival rate of 79.7%. The present research indicates that EM is a challenging malignancy, but has a relatively better prognosis and easier management than other melanomas of the head and neck region. These are probably related to the anatomical location which leads to early diagnosis. Therefore, EM should be considered as a specific disease requiring dedicated treatment. Based on the personal authors' experience and comprehensive overview of the current knowledge, a dedicated protocol is proposed.
Collapse
Affiliation(s)
- Federica Dini
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, Florence
| | - Pietro Susini
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | | | - Giuseppe Nisi
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | - Roberto Cuomo
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | - Luca Grimaldi
- Plastic Surgery Unit, Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena
| | | | - Luca Tinunin
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence
| | - Pietro Antonini
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona
| | | | | | | | - Laura Doni
- Clinical Oncology Unit, Careggi University Hospital
| | - Cinzia Mazzini
- Unit of Ocular Oncology, Department of Neuromuscular and Sense Organs
| | - Nicola Santoro
- Unit of Ocular Oncology, Department of Surgery and Translational Medicine, Careggi University Hospital, Florence, Italy
| | | |
Collapse
|
296
|
Tocchetti CG, Farmakis D, Koop Y, Andres MS, Couch LS, Formisano L, Ciardiello F, Pane F, Au L, Emmerich M, Plummer C, Gulati G, Ramalingam S, Cardinale D, Brezden-Masley C, Iakobishvili Z, Thavendiranathan P, Santoro C, Bergler-Klein J, Keramida K, de Boer RA, Maack C, Lutgens E, Rassaf T, Fradley MG, Moslehi J, Yang EH, De Keulenaer G, Ameri P, Bax J, Neilan TG, Herrmann J, Mbakwem AC, Mirabel M, Skouri H, Hirsch E, Cohen-Solal A, Sverdlov AL, van der Meer P, Asteggiano R, Barac A, Ky B, Lenihan D, Dent S, Seferovic P, Coats AJS, Metra M, Rosano G, Suter T, Lopez-Fernandez T, Lyon AR. Cardiovascular toxicities of immune therapies for cancer - a scientific statement of the Heart Failure Association (HFA) of the ESC and the ESC Council of Cardio-Oncology. Eur J Heart Fail 2024; 26:2055-2076. [PMID: 39087551 DOI: 10.1002/ejhf.3340] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
The advent of immunological therapies has revolutionized the treatment of solid and haematological cancers over the last decade. Licensed therapies which activate the immune system to target cancer cells can be broadly divided into two classes. The first class are antibodies that inhibit immune checkpoint signalling, known as immune checkpoint inhibitors (ICIs). The second class are cell-based immune therapies including chimeric antigen receptor T lymphocyte (CAR-T) cell therapies, natural killer (NK) cell therapies, and tumour infiltrating lymphocyte (TIL) therapies. The clinical efficacy of all these treatments generally outweighs the risks, but there is a high rate of immune-related adverse events (irAEs), which are often unpredictable in timing with clinical sequalae ranging from mild (e.g. rash) to severe or even fatal (e.g. myocarditis, cytokine release syndrome) and reversible to permanent (e.g. endocrinopathies).The mechanisms underpinning irAE pathology vary across different irAE complications and syndromes, reflecting the broad clinical phenotypes observed and the variability of different individual immune responses, and are poorly understood overall. Immune-related cardiovascular toxicities have emerged, and our understanding has evolved from focussing initially on rare but fatal ICI-related myocarditis with cardiogenic shock to more common complications including less severe ICI-related myocarditis, pericarditis, arrhythmias, including conduction system disease and heart block, non-inflammatory heart failure, takotsubo syndrome and coronary artery disease. In this scientific statement on the cardiovascular toxicities of immune therapies for cancer, we summarize the pathophysiology, epidemiology, diagnosis, and management of ICI, CAR-T, NK, and TIL therapies. We also highlight gaps in the literature and where future research should focus.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Dimitrios Farmakis
- Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Yvonne Koop
- Department of Cardiovascular Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Dutch Heart Foundation, The Hague, The Netherlands
| | - Maria Sol Andres
- Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Liam S Couch
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Lewis Au
- Skin and Renal Unit, Royal Marsden NHS Foundation Trust, London, UK
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Max Emmerich
- The Francis Crick Institute, London, UK
- The Royal Marsden Hospital, London, UK
- St. John's Institute of Dermatology, Guy's and St Thomas' Hospital, London, UK
| | - Chris Plummer
- Department of Cardiology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Geeta Gulati
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Oslo University Hospital, Ullevål, Oslo, Norway
- Division of Research and Innovation, Akershus University Hospital, Lørenskog, Norway
| | - Sivatharshini Ramalingam
- Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
- Oxted Health Centre, Oxted, UK
| | - Daniela Cardinale
- CardioOncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Zaza Iakobishvili
- Department of Community Cardiology, Tel Aviv Jaffa District, Clalit Health Services, Tel Aviv, Israel
- Department of Cardiology, Assuta Ashdod University Hospital, Ashdod, Israel
- Faculty of Health Sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Ciro Santoro
- Department of Advanced Biomedical Science, Federico II University Hospital, Naples, Italy
| | | | - Kalliopi Keramida
- Cardiology Department, General Anti-Cancer, Oncological Hospital, Agios Savvas, Athens, Greece
| | - Rudolf A de Boer
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
- Medical Clinic 1, University Clinic Würzburg, Würzburg, Germany
| | - Esther Lutgens
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Javid Moslehi
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gilles De Keulenaer
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Jeroen Bax
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joerg Herrmann
- Cardio-Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amam C Mbakwem
- College of Medicine, University of Lagos/Lagos University Teaching Hospital Idi Araba, Lagos, Nigeria
| | | | - Hadi Skouri
- Cardiology Division, Sheikh Shakhbout Medical City, Khalifa University, Abu Dhabi, UAE
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Alain Cohen-Solal
- Paris Cité University INSERM U 948 MASCOT Research Unit Cardiology, Lariboisere Universitaire Hospital, AP-HP, Paris, France
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, University of Newcastle, Hunter Medical Research Institute, Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Riccardo Asteggiano
- Internal Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
- LARC, Laboratorio Analisi e Ricerca Clinica, Turin, Italy
| | - Ana Barac
- Inova Schar Heart and Vascular Institute, Falls Church, VA, USA
| | - Bonnie Ky
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Lenihan
- Cape Cardiology, St Francis Healthcare, Cape Girardeau, MO, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, USA
| | - Petar Seferovic
- University Medical Center, Medical Faculty University of Belgrade, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | | | - Marco Metra
- Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppe Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, FR, Italy
| | - Thomas Suter
- Bern University Hospital, University of Bern, Bern, Switzerland
| | - Teresa Lopez-Fernandez
- Cardio-Oncology Unit, Cardiology Department, IdiPAZ Research Institute, La Paz University Hospital, Madrid, Spain
- Cardiology Department, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
297
|
Taglialatela I, Indini A, Santanelli G, Di Liberti G, Di Guardo L, De Braud F, Del Vecchio M. Melanoma and sex hormones: Pathogenesis, progressive disease and response to treatments. TUMORI JOURNAL 2024; 110:309-318. [PMID: 38372040 DOI: 10.1177/03008916241231687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Cutaneous melanoma represents the fifth tumor in terms of incidence in young adults, with a major involvement of males than females. Despite the significant changes in available effective treatments for cutaneous melanoma, there is still a proportion of patients that do not benefit long-term disease control with immune checkpoint inhibitors and/or BRAF/MEK inhibitors, and eventually develop progressive disease. In addition to the emerging biomarkers under investigation to understand resistance to treatments, recent studies resumed the role of sex hormones (estrogens, progesterone and androgens) in melanoma patients. In the last decades, the impact of sex hormones has been considered controversial in melanoma patients, but actual growing preclinical and clinical evidence underline the potential influence on melanoma cells' growth, tumor microenvironment, the immune system and consequently on the course of disease.This review will provide available insights on the role of sex hormones in melanoma pathogenesis, disease progression and response/resistance to systemic treatments. We will also offer an overview on the recent studies on the theme, describing the hormonal contribution to disease response and the interaction with targeted therapies and immune-checkpoint inhibitors in cutaneous melanoma patients, illustrating an insight into future research in this field.
Collapse
Affiliation(s)
- Ida Taglialatela
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Alice Indini
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giulia Santanelli
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giorgia Di Liberti
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Lorenza Di Guardo
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Filippo De Braud
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
- Università degli studi di Milano, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Michele Del Vecchio
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
298
|
Narvaez-Rojas AR, Linhares S, Sedighim S, Klingbeil KD, Milikowski C, Elgart G, Jaimes N, Feun L, Lutzky J, De la Cruz Ku G, Avisar E, Möller MG. Is primary breast melanoma a true pathological entity? The argument against it. Heliyon 2024; 10:e37224. [PMID: 39309840 PMCID: PMC11414497 DOI: 10.1016/j.heliyon.2024.e37224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Background Previous studies have reported cases of primary melanoma of the breast parenchyma (PMBP), but the pathogenesis of this disease remains poorly understood. We review the presentation and outcomes of reported cases and provide detailed pathological analysis of four additional cases. Furthermore, we discuss potential theories regarding the pathogenesis of this clinical presentation. Results We identified 29 published studies (n = 95 patients) and report four new cases (n = 99). Ninety-one (92 %) patients were female, with a median age of 50 years. Previous skin melanomas were reported by 56 % of patients, with the trunk being the most common location (32.7 %) followed by the upper extremities (20 %). The most common tumor location reported (n = 73) was the right (49 %) upper outer quadrant (56 %). The median time from skin melanoma diagnosis to the presence of a breast mass was 65 months (1-192). Nodal status at presentation was reported in n = 67 (68 %) patients. Of these, positive nodal metastases were seen in 40.3 %, while distant metastatic disease at presentation was reported in 30 % of patients. Surgery was performed in 66 %, being partial mastectomy (PM) the most common procedure in 82 %. Adjuvant therapy was described in 38 patients. The reported (n = 12) median survival was 11.5 (1-70) months. Conclusion Melanomas identified in the breast parenchyma are likely the result of nodal or hematogenous spread from previously known or unknown melanomas, and should not be considered as PMBP. Management should be multidisciplinary, including surgical excision aimed at obtaining negative margins with lymphadenectomy of clinically positive nodes and neoadjuvant/adjuvant immunotherapy.
Collapse
Affiliation(s)
- Alexis R. Narvaez-Rojas
- Department of Radiation Oncology, Maimonides Cancer Center, Brooklyn, NY, USA
- International Coalition on Surgical Research, Universidad Nacional Autónoma de Nicaragua, UNAN, Managua, Nicaragua
| | | | - Shaina Sedighim
- Department of Surgery, University of California, Irvine, Orange, CA, USA
| | - Kyle Daniel Klingbeil
- Department of Surgery, University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Clara Milikowski
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Pathology, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - George Elgart
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Dermatology, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Natalia Jaimes
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Dermatology, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lynn Feun
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Medicine, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jose Lutzky
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Medicine, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Eli Avisar
- University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, Division of Surgical Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mecker G. Möller
- University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, Division of Surgical Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
299
|
Claeys A, Van den Eynden J. MHC class II genotypes are independent predictors of anti-PD1 immunotherapy response in melanoma. COMMUNICATIONS MEDICINE 2024; 4:184. [PMID: 39349759 PMCID: PMC11443121 DOI: 10.1038/s43856-024-00612-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Immune checkpoint blockade is a highly successful anti-cancer immunotherapy. Both CTLA4 and PD1 checkpoint blockers are clinically available for melanoma treatment, with anti-PD1 therapy reaching response rates of 35-40%. These responses, which are mediated via neoantigen presentation by the polymorphic MHC complex, are hard to predict and the tumor mutation burden is currently one of the few available biomarkers. While MHC genotypes are expected to determine therapy responses, association studies have remained largely elusive. METHODS We developed an overall MHC genotype binding score (MGBS), indicative of a patient's MHC class I (MHC-I) and class II (MHC-II) neoantigen binding capacity and solely based on the germline MHC-I (MGBS-I) and MHC-II (MGBS-II) genotypes. These scores were then correlated to survival and clinical responses following anti-PD1 immunotherapy in a previously published dataset of 144 melanoma patients. RESULTS We demonstrate that MGBS scores are TMB-independent predictors of anti-PD1 immunotherapy responses in melanoma. Opposite outcomes were found for both MHC classes, with high MGBS-I and MGBS-II predicting good and bad outcomes, respectively. Interestingly, high MGBS-II is mainly associated with treatment response failure in a subgroup of anti-CTLA4 pretreated patients. CONCLUSIONS Our results suggest that MGBS, calculated solely from the MHC genotype, has clinical potential as a non-invasive and tumor-independent biomarker to guide anti-cancer immunotherapy in melanoma.
Collapse
Affiliation(s)
- Arne Claeys
- Department of Human Structure and Repair, Unit of Anatomy and Embryology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Unit of Anatomy and Embryology, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
300
|
Nader NE, Frederico SC, Miller T, Huq S, Zhang X, Kohanbash G, Hadjipanayis CG. Barriers to T Cell Functionality in the Glioblastoma Microenvironment. Cancers (Basel) 2024; 16:3273. [PMID: 39409893 PMCID: PMC11476085 DOI: 10.3390/cancers16193273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor depicted by a cold tumor microenvironment, low immunogenicity, and limited effective therapeutic interventions. Its location in the brain, a highly immune-selective organ, acts as a barrier, limiting immune access and promoting GBM dissemination, despite therapeutic interventions. Currently, chemotherapy and radiation combined with surgical resection are the standard of care for GBM treatment. Although immune checkpoint blockade has revolutionized the treatment of solid tumors, its observed success in extracranial tumors has not translated into a significant survival benefit for GBM patients. To develop effective immunotherapies for GBM, it is vital to tailor treatments to overcome the numerous immunosuppressive barriers that inhibit T cell responses to these tumors. In this review, we address the unique physical and immunological barriers that make GBM challenging to treat. Additionally, we explore potential therapeutic mechanisms, studied in central nervous system (CNS) and non-CNS cancers, that may overcome these barriers. Furthermore, we examine current and promising immunotherapy clinical trials and immunotherapeutic interventions for GBM. By highlighting the array of challenges T cell-based therapies face in GBM, we hope this review can guide investigators as they develop future immunotherapies for this highly aggressive malignancy.
Collapse
Affiliation(s)
- Noor E. Nader
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (N.E.N.); (S.C.F.); (T.M.)
| | - Stephen C. Frederico
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (N.E.N.); (S.C.F.); (T.M.)
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Tracy Miller
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (N.E.N.); (S.C.F.); (T.M.)
| | - Sakibul Huq
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Xiaoran Zhang
- Sloan Kettering Memorial Cancer Center, New York, NY 10065, USA;
| | - Gary Kohanbash
- Sloan Kettering Memorial Cancer Center, New York, NY 10065, USA;
| | | |
Collapse
|