351
|
Evans CR. Overcoming combination fatigue: Addressing high-dimensional effect measure modification and interaction in clinical, biomedical, and epidemiologic research using multilevel analysis of individual heterogeneity and discriminatory accuracy (MAIHDA). Soc Sci Med 2024; 340:116493. [PMID: 38128257 DOI: 10.1016/j.socscimed.2023.116493] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Growing interest in precision medicine, gene-environment interactions, health equity, expanding diversity in research, and the generalizability results, requires researchers to evaluate how the effects of treatments or exposures differ across numerous subgroups. Evaluating combination complexity, in the form of effect measure modification and interaction, is therefore a common study aim in the biomedical, clinical, and epidemiologic sciences. There is also substantial interest in expanding the combinations of factors analyzed to include complex treatment protocols (e.g., multiple study arms or factorial randomization), comorbid medical conditions or risk factors, and sociodemographic and other subgroup identifiers. However, expanding the number of subgroup category combinations creates combination fatigue problems, including concerns over small sample size, reduced power, multiple testing, spurious results, and design and analytic complexity. Creative new approaches for managing combination fatigue and evaluating high-dimensional effect measure modification and interaction are needed. Intersectional MAIHDA (multilevel analysis of individual heterogeneity and discriminatory accuracy) has already attracted substantial interest in social epidemiology, and has been hailed as the new gold standard for investigating health inequities across complex intersections of social identity. Leveraging the inherent advantages of multilevel models, a more general multicategorical MAIHDA can be used to study statistical interactions and predict effects across high-dimensional combinations of conditions, with important advantages over alternative approaches. Though it has primarily been used thus far as an analytic approach, MAIHDA should also be used as a framework for study design. In this article, I introduce MAIHDA to the broader health sciences research community, discuss its advantages over conventional approaches, and provide an overview of potential applications in clinical, biomedical, and epidemiologic research.
Collapse
Affiliation(s)
- Clare R Evans
- Department of Sociology, 1291 University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
352
|
van Akkooi ACJ. Relatlimab, an Immune Checkpoint Inhibitor that Blocks LAG-3, the Latest Drug to be Added to the Arsenal of Systemic Therapies for Melanoma: What Does a Surgical Oncologist Need to Know? Ann Surg Oncol 2024; 31:1-3. [PMID: 37843663 DOI: 10.1245/s10434-023-14416-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023]
Affiliation(s)
- Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
353
|
Poletto S, Paruzzo L, Nepote A, Caravelli D, Sangiolo D, Carnevale-Schianca F. Predictive Factors in Metastatic Melanoma Treated with Immune Checkpoint Inhibitors: From Clinical Practice to Future Perspective. Cancers (Basel) 2023; 16:101. [PMID: 38201531 PMCID: PMC10778365 DOI: 10.3390/cancers16010101] [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: 10/10/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
The introduction of immunotherapy revolutionized the treatment landscape in metastatic melanoma. Despite the impressive results associated with immune checkpoint inhibitors (ICIs), only a portion of patients obtain a response to this treatment. In this scenario, the research of predictive factors is fundamental to identify patients who may have a response and to exclude patients with a low possibility to respond. These factors can be host-associated, immune system activation-related, and tumor-related. Patient-related factors can vary from data obtained by medical history (performance status, age, sex, body mass index, concomitant medications, and comorbidities) to analysis of the gut microbiome from fecal samples. Tumor-related factors can reflect tumor burden (metastatic sites, lactate dehydrogenase, C-reactive protein, and circulating tumor DNA) or can derive from the analysis of tumor samples (driver mutations, tumor-infiltrating lymphocytes, and myeloid cells). Biomarkers evaluating the immune system activation, such as IFN-gamma gene expression profile and analysis of circulating immune cell subsets, have emerged in recent years as significantly correlated with response to ICIs. In this manuscript, we critically reviewed the most updated literature data on the landscape of predictive factors in metastatic melanoma treated with ICIs. We focus on the principal limits and potentiality of different methods, shedding light on the more promising biomarkers.
Collapse
Affiliation(s)
- Stefano Poletto
- Department of Oncology, University of Turin, AOU S. Luigi Gonzaga, 10043 Orbassano, Italy;
| | - Luca Paruzzo
- Department of Oncology, University of Turin, 10124 Turin, Italy; (L.P.); (D.S.)
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alessandro Nepote
- Department of Oncology, University of Turin, AOU S. Luigi Gonzaga, 10043 Orbassano, Italy;
| | - Daniela Caravelli
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCs, 10060 Candiolo, Italy; (D.C.); (F.C.-S.)
| | - Dario Sangiolo
- Department of Oncology, University of Turin, 10124 Turin, Italy; (L.P.); (D.S.)
| | | |
Collapse
|
354
|
Amylidi AL, Gogadis A, Yerolatsite M, Zarkavelis G, Torounidou N, Keramisanou V, Kampletsas E, Mauri D. Exploring a Rarity: Incidence of and Therapeutic Approaches for Neurological Complications and Hypophysitis in Cancer Patients on Immune Checkpoint Inhibitors-A Single-Center Study. Curr Oncol 2023; 30:10509-10518. [PMID: 38132400 PMCID: PMC10742161 DOI: 10.3390/curroncol30120766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 inhibitors, have become the standard of care for many cancer types. However, they induce immune-related adverse events (irAEs), including neurotoxicity and hypophysitis. The incidence and outcomes of neurotoxicity and hypophysitis in patients treated with immune checkpoint inhibitors are not well established. We conducted a retrospective study of 812 patients with solid cancers who received immune checkpoint inhibitors at the University General Hospital of Ioannina between January 2018 and January 2023. We assessed demographic and clinical data, including the severity of symptoms, treatment regimen, other irAEs, resolution type and time, and death. Two patients experienced neurotoxicity and two hypophysitis. All four patients required inpatient administration and received corticosteroids or/and hormone replacement. Three patients responded to the initial therapy, experiencing full recovery, while one patient was corticosteroid-resistant, and immunoglobin G was administered. Two patients never received immunotherapy after their toxicity due to the severity of symptoms; one patient continued monotherapy with nivolumab, changing from combination therapy with ipilimumab-nivolumab, while the fourth patient continued his initial treatment with nivolumab. Our study suggests that the incidence of neurotoxicity and hypophysitis in patients treated with immune checkpoint inhibitors is low, but careful monitoring and prompt treatment with corticosteroids are necessary for effective management.
Collapse
Affiliation(s)
- Anna Lea Amylidi
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Aristeidis Gogadis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Melina Yerolatsite
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Nanteznta Torounidou
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Varvara Keramisanou
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Eleftherios Kampletsas
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Davide Mauri
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (A.G.); (M.Y.); (G.Z.); (N.T.); (V.K.); (E.K.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
355
|
Kibel S, Kuehne N, Ribeiro MF, Muniz TP, Ye XY, Spreafico A, Saibil SD, Sun A, Mak DY, Gray D, Jones B, Wong P, Butler MO. The Role of Adjuvant Radiotherapy for the Treatment of Resected High-Risk Stage III Cutaneous Melanoma in the Era of Modern Systemic Therapies. Cancers (Basel) 2023; 15:5867. [PMID: 38136412 PMCID: PMC10741555 DOI: 10.3390/cancers15245867] [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: 11/12/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Modern adjuvant systemic therapies (STs) have revolutionized the management of stage III melanoma. Currently, the role of adjuvant radiotherapy (RT) remains unclear. In this single-center retrospective study, patients with clinically detectable stage III melanoma with high-risk features for lymph node basin (LNB) recurrence and whose tumors were fully resected with complete lymphadenectomy (CLD) between 2010 and 2019 were assessed. We determined the cumulative incidence (CIF) of LNB recurrence and any disease recurrence or progression using competing risk analysis. A total of 108 patients were identified; the median age was 59 years (24-92), and 74 (69%) were men. A total of 51 (42%) received adjuvant RT, 22 (20%) received adjuvant ST, and 35 (32%) received no adjuvant therapy. The advent of ST changed clinical practice, with a significant increase in the use of adjuvant ST and a decrease in the use of RT when comparing practice patterns before and after 2015 (p < 0.001). The 3-year CIF of LNB recurrence was similar in patients treated with adjuvant RT (6.3%) and adjuvant ST (9.8%). The 3-year CIF of any disease recurrence or progression was lower in patients receiving adjuvant ST (24%) compared to those receiving adjuvant RT (52%) or no adjuvant therapy (55%, p = 0.06). Three-year overall survival (OS) was not significantly different in patients treated with ST compared to those not treated with any ST (p = 0.118). Despite ST replacing RT as the dominant adjuvant treatment modality, this change in practice has not resulted in increased LNB recurrence for patients at high risk of LNB recurrence following CLD.
Collapse
Affiliation(s)
- Seth Kibel
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada; (S.K.); (N.K.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Nathan Kuehne
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada; (S.K.); (N.K.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Mauricio Fernando Ribeiro
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Thiago P. Muniz
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Xiang Y. Ye
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Anna Spreafico
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada; (S.K.); (N.K.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Samuel D. Saibil
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada; (S.K.); (N.K.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Alexander Sun
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - David Y. Mak
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Diana Gray
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Bailie Jones
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Philip Wong
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Marcus O. Butler
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada; (S.K.); (N.K.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
356
|
Yu C, Hsieh K, Cherry DR, Nehlsen AD, Resende Salgado L, Lazarev S, Sindhu KK. Immune Escape in Glioblastoma: Mechanisms of Action and Implications for Immune Checkpoint Inhibitors and CAR T-Cell Therapy. BIOLOGY 2023; 12:1528. [PMID: 38132354 PMCID: PMC10741174 DOI: 10.3390/biology12121528] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Glioblastoma, the most common primary brain cancer in adults, is characterized by a poor prognosis and resistance to standard treatments. The advent of immunotherapy has revolutionized the treatment of several cancers in recent years but has failed to demonstrate benefit in patients with glioblastoma. Understanding the mechanisms by which glioblastoma exerts tumor-mediated immune suppression in both the tumor microenvironment and the systemic immune landscape is a critical step towards developing effective immunotherapeutic strategies. In this review, we discuss the current understanding of immune escape mechanisms in glioblastoma that compromise the efficacy of immunotherapies, with an emphasis on immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy. In parallel, we review data from preclinical studies that have identified additional therapeutic targets that may enhance overall treatment efficacy in glioblastoma when administered alongside existing immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kunal K. Sindhu
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.Y.); (D.R.C.); (A.D.N.); (L.R.S.); (S.L.)
| |
Collapse
|
357
|
Benito JM, Restrepo C, García-Foncillas J, Rallón N. Immune checkpoint inhibitors as potential therapy for reverting T-cell exhaustion and reverting HIV latency in people living with HIV. Front Immunol 2023; 14:1270881. [PMID: 38130714 PMCID: PMC10733458 DOI: 10.3389/fimmu.2023.1270881] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
The immune system of people living with HIV (PLWH) is persistently exposed to antigens leading to systemic inflammation despite combination antiretroviral treatment (cART). This inflammatory milieu promotes T-cell activation and exhaustion. Furthermore, it produces diminished effector functions including loss of cytokine production, cytotoxicity, and proliferation, leading to disease progression. Exhausted T cells show overexpression of immune checkpoint molecules (ICs) on the cell surface, including programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), and lymphocyte activation gene-3 (LAG-3). The ICs also play a crucial role in T-cell exhaustion by reducing the immune response to cancer antigens. Immunotherapy based on immune checkpoint inhibitors (ICIs) has changed the management of a diversity of cancers. Additionally, the interest in exploring this approach in the setting of HIV infection has increased, including AIDS-defining cancers and non-AIDS-defining cancers in PLWH. To date, research on this topic suggests that ICI-based therapies in PLWH could be a safe and effective approach. In this review, we provide an overview of the current literature on the potential role of ICI-based immunotherapy not only in cancer remission in PLWH but also as a therapeutic intervention to restore immune response against HIV, revert HIV latency, and attain a functional cure for HIV infection.
Collapse
Affiliation(s)
- José M. Benito
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | | | - Jesús García-Foncillas
- Department of Oncology and Cancer Institute, Fundacion Jimenez Diaz University Hospital, Autonomous University, Madrid, Spain
| | - Norma Rallón
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| |
Collapse
|
358
|
Green CE, Chacon J, Godinich BM, Hock R, Kiesewetter M, Raynor M, Marwaha K, Maharaj S, Holland N. The Heart of the Matter: Immune Checkpoint Inhibitors and Immune-Related Adverse Events on the Cardiovascular System. Cancers (Basel) 2023; 15:5707. [PMID: 38136253 PMCID: PMC10742007 DOI: 10.3390/cancers15245707] [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: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer remains a prominent global cause of mortality, second only to cardiovascular disease. The past decades have witnessed substantial advancements in anti-cancer therapies, resulting in improved outcomes. Among these advancements, immunotherapy has emerged as a promising breakthrough, leveraging the immune system to target and eliminate cancer cells. Despite the remarkable potential of immunotherapy, concerns have arisen regarding associations with adverse cardiovascular events. This review examines the complex interplay between immunotherapy and cardiovascular toxicity and provides an overview of immunotherapy mechanisms, clinical perspectives, and potential biomarkers for adverse events, while delving into the intricate immune responses and evasion mechanisms displayed by cancer cells. The focus extends to the role of immune checkpoint inhibitors in cancer therapy, including CTLA-4, PD-1, and PD-L1 targeting antibodies. This review underscores the multifaceted challenges of managing immunotherapy-related cardiovascular toxicity. Risk factors for immune-related adverse events and major adverse cardiac events are explored, encompassing pharmacological, treatment-related, autoimmune, cardiovascular, tumor-related, social, genetic, and immune-related factors. The review also advocates for enhanced medical education and risk assessment tools to identify high-risk patients for preventive measures. Baseline cardiovascular evaluations, potential prophylactic strategies, and monitoring of emerging toxicity symptoms are discussed, along with the potential of adjunct anti-inflammatory therapies.
Collapse
Affiliation(s)
- Chase E. Green
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Jessica Chacon
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Brandon M. Godinich
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Rivers Hock
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Maria Kiesewetter
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Mark Raynor
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Komal Marwaha
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Satish Maharaj
- Department of Internal Medicine, Division of Hematology/Oncology, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 4800 Alberta Ave., El Paso, TX 79905, USA
| | - Nathan Holland
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| |
Collapse
|
359
|
Boutros A, Carosio R, Campanella D, Spagnolo F, Banelli B, Morabito A, Pistillo M, Croce E, Cecchi F, Pronzato P, Queirolo P, Raposio E, Fontana V, Tanda E. The predictive and prognostic role of single nucleotide gene variants of PD-1 and PD-L1 in patients with advanced melanoma treated with PD-1 inhibitors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100408. [PMID: 38192613 PMCID: PMC10772261 DOI: 10.1016/j.iotech.2023.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Background Despite having revolutionized the treatment paradigm for advanced melanoma, not all patients benefit from immune checkpoint inhibitor therapy. To date, there are no predictive biomarkers for response or the occurrence of immune-related adverse events (irAEs) to programmed cell death protein 1 (PD-1) inhibitors. Our aim was to investigate the predictive and prognostic role of single nucleotide variants (SNVs) of genes involved in the PD-1 axis. Methods We analysed, in metastatic melanoma patients treated with nivolumab or pembrolizumab, five PD-1 SNVs, namely PD1.3 G>A (rs11568821), PD1.5 C>T (rs2227981), PD1.6 G>A (rs10204525), PD1.7 T>C(rs7421861), PD1.10 C>G (rs5582977) and three programmed death-ligand 1 (PD-L1) SNVs: +8293 C>A (rs2890658), PD-L1 C>T (rs2297136) and PD-L1 G>C (rs4143815). Association of SNV genotypic frequencies with best overall response to PD-1 inhibitors and development of irAEs were estimated through a modified Poisson regression. A Cox regression modelling approach was applied to evaluate the SNV association with OS. Results A total of 125 patients with advanced melanoma were included in the analysis. A reduction in irAEs risk was observed in patients carrying the PD-L1 +8293 C/A genotype compared with those carrying the C/C genotype (risk ratio = 0.45; 95% CL 0.22-0.93; P = 0.031). A trend for a reduction in irAEs was also observed with the PD1.5 T allele (risk ratio = 0.70, 95% confidence limits 0.48-1.01 versus C allele). None of the SNVs was associated with response to therapy. Finally, a survival benefit was observed in patients harbouring the PD1.7 C/C genotype (hazard ratio = 0.37; 95% confidence limits 0.14-0.96; P = 0.028) in the homozygous model. Conclusions Our study showed that PD-1.5 and PD-L1 +8293 SNVs may play a role as a predictive biomarker of development of irAEs to PD-1 inhibitors. PD1.7 SNV may also be associated with a reduction of the risk of death, although further translational research is needed to confirm these results.
Collapse
Affiliation(s)
- A. Boutros
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa
| | - R. Carosio
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - D. Campanella
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - F. Spagnolo
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genoa
| | - B. Banelli
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - A. Morabito
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - M.P. Pistillo
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - E. Croce
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa
| | - F. Cecchi
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| | - P. Pronzato
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| | - P. Queirolo
- Division of Melanoma Sarcoma and Rare Tumors, IRCCS European Institute of Oncology, Milan, Italy
| | - E. Raposio
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genoa
| | - V. Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - E.T. Tanda
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| |
Collapse
|
360
|
Fu J, Yao F, An Y, Li X, Wang W, Yang XD. Novel bispecific aptamer targeting PD-1 and nucleolin for cancer immunotherapy. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00177-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Abstract
Background
Immune checkpoint blockade (ICB) is a promising strategy for cancer treatment and has achieved remarkable clinical results. Further improvement of ICB efficacy may advance cancer immunotherapy and has evident medical importance. Here in this study, a PD-1 aptamer was functionalized with a tumor-homing nucleolin aptamer (AS1411) to build a novel bispecific agent (BiApt) for boosting the efficacy of ICB therapy.
Results
The two aptamers were coupled together via sticky ends to form BiApt, which had an average size of 11.70 nm. Flow cytometry revealed that BiApt could bind with both the activated T cells and the nucleolin-expressing tumor cells. In addition, BiApt could recruit more T cells to the vicinity of nucleolin-positive tumor cells. Functionally, BiApt enhanced the PBMC-mediated anticancer cytotoxicity in vitro compared with free PD-1 aptamer. Moreover, in an animal model of CT26 colon cancer, BiApt significantly boosted the antitumor efficacy vs. free PD-1 aptamer.
Conclusion
The results suggest that bispecific agent combining ICB and tumor-homing functions has potential to improve the efficacy of ICB immunotherapy.
Collapse
|
361
|
Gelmi MC, Gezgin G, van der Velden PA, Luyten GPM, Luk SJ, Heemskerk MHM, Jager MJ. PRAME Expression: A Target for Cancer Immunotherapy and a Prognostic Factor in Uveal Melanoma. Invest Ophthalmol Vis Sci 2023; 64:36. [PMID: 38149971 PMCID: PMC10755595 DOI: 10.1167/iovs.64.15.36] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/15/2023] [Indexed: 12/28/2023] Open
Abstract
Purpose Uveal melanoma (UM) is a rare disease with a high mortality, and new therapeutic options are being investigated. Preferentially Expressed Antigen in Melanoma (PRAME) is a cancer testis antigen, expressed in the testis, but also in cancers, including uveal melanoma. PRAME is considered a target for immune therapy in several cancers, and PRAME-specific T cell clones have been shown to kill UM cells. Methods We studied the literature on PRAME expression in hematological and solid malignancies, including UM, and its role as a target for immunotherapy. The distribution of tumor features was compared between PRAME-high and PRAME-low UM in a 64-patient cohort from the Leiden University Medical Center (LUMC) and in the Cancer Genome Atlas (TCGA) cohort of 80 cases and differential gene expression analysis was performed in the LUMC cohort. Results PRAME is expressed in many malignancies, it is frequently associated with a negative prognosis, and can be the target of T cell receptor (TCR)-transduced T cells, a promising treatment option with high avidity and safety. In UM, PRAME is expressed in 26% to 45% of cases and is correlated with a worse prognosis. In the LUMC and the TCGA cohorts, high PRAME expression was associated with larger diameter, higher Tumor-Node-Metastasis (TNM) stage, more frequent gain of chromosome 8q, and an inflammatory phenotype. Conclusions We confirm that PRAME is associated with poor prognosis in UM and has a strong connection with extra copies of 8q. We show that PRAME-specific immunotherapy in an adjuvant setting is promising in treatment of malignancies, including UM.
Collapse
Affiliation(s)
- Maria Chiara Gelmi
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gulçin Gezgin
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Sietse J. Luk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
362
|
Schina A, Pedersen S, Spenning AL, Laursen OK, Pedersen C, Haslund CA, Schmidt H, Bastholt L, Svane IM, Ellebaek E, Donia M. Sustained improved survival of patients with metastatic melanoma after the introduction of anti-PD-1-based therapies. Eur J Cancer 2023; 195:113392. [PMID: 37924648 DOI: 10.1016/j.ejca.2023.113392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND The introduction of modern therapies improved the median survival of patients with metastatic melanoma (MM). Here, we determined the real-world impact of modern treatments on the long-term survival of MM. METHODS In a population-based study, we extracted all cases of MM diagnosed in four non-consecutive years marked by major changes in available 1st line treatments (2012, 2014, 2016, and 2018) from the Danish MM Database. Patients were grouped into "trial-like" and "trial-excluded" based on common trial eligibility criteria. RESULTS We observed a sustained improved survival of "trial-like" patients diagnosed in 2016 or in 2018, compared to 2012 or 2014, but no major differences in 2018 versus 2016. In contrast, while survival of "trial-excluded" patients in 2016 was better compared to 2014 and 2012, survival in 2018 was improved over all previous years. We then developed a prognostic model based on multivariable stratified Cox regression, to predict the survival of newly diagnosed MM patients. Internal validation showed excellent discrimination and calibration, with a time-area-under-the-curve above 0.79 at multiple time horizons, for up to four years after diagnosis. CONCLUSIONS The introduction of modern treatments such as anti-PD-1 has led to a sustained, improved survival of real-world patients with MM, regardless of their eligibility for clinical trials. We provide an updateable prognostic model that can be used to improve patient information. Overall, these data highlight a positive population-based impact of modern treatments and can help health technology assessment agencies worldwide to evaluate the appropriateness of drug pricing based on known cost-benefit data.
Collapse
Affiliation(s)
- Aimilia Schina
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Sidsel Pedersen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | | | - Cecilia Pedersen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| | - Marco Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| |
Collapse
|
363
|
Alvarez J, Smith JJ. Anorectal mucosal melanoma. SEMINARS IN COLON AND RECTAL SURGERY 2023; 34:100990. [PMID: 38746826 PMCID: PMC11090490 DOI: 10.1016/j.scrs.2023.100990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Anorectal mucosal melanoma accounts for less than 1 % of all anorectal malignant tumors and a tendency for delayed diagnosis leads to advanced disease at presentation.1,2 Due to the rarity of the disease, there are limited prospective trials exploring the optimal treatment strategies. Generally, tumors are surgically excised, with a preference for conservative management with wide local excision. In the past decade, there have been advances with immunotherapy and other targeted therapies. Multiple clinical trials continue exploring neoadjuvant/adjuvant combination treatments in the setting of advanced or unresectable disease.
Collapse
Affiliation(s)
- Janet Alvarez
- Research Scholar, Memorial Sloan Kettering Cancer Center, Department of Surgery, 1275 York Avenue | SR-201, New York, NY 10065, USA
| | - J. Joshua Smith
- Associate Member, Associate Attending Surgeon, Memorial Sloan Kettering Cancer Center, Colorectal Service, Department of Surgery, 1275 York Avenue, SR-201, New York, NY 10065, USA
| |
Collapse
|
364
|
Reticker-Flynn NE, Engleman EG. Lymph nodes: at the intersection of cancer treatment and progression. Trends Cell Biol 2023; 33:1021-1034. [PMID: 37149414 PMCID: PMC10624650 DOI: 10.1016/j.tcb.2023.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
Metastasis to lymph nodes (LNs) is a common feature of disease progression in most solid organ malignancies. Consequently, LN biopsy and lymphadenectomy are common clinical practices, not only because of their diagnostic utility but also as a means of deterring further metastatic spread. LN metastases have the potential to seed additional tissues and can induce metastatic tolerance, a process by which tumor-specific immune tolerance in LNs promotes further disease progression. Nonetheless, phylogenetic studies have revealed that distant metastases are not necessarily derived from nodal metastases. Furthermore, immunotherapy efficacy is increasingly being attributed to initiation of systemic immune responses within LNs. We argue that lymphadenectomy and nodal irradiation should be approached with caution, particularly in patients receiving immunotherapy.
Collapse
Affiliation(s)
- Nathan E Reticker-Flynn
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
365
|
Rocha GIY, Gomes JEM, Leite ML, da Cunha NB, Costa FF. Epigenome-Driven Strategies for Personalized Cancer Immunotherapy. Cancer Manag Res 2023; 15:1351-1367. [PMID: 38058537 PMCID: PMC10697012 DOI: 10.2147/cmar.s272031] [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: 05/13/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
Fighting cancer remains one of the greatest challenges for science in the 21st century. Advances in immunotherapy against different types of cancer have greatly contributed to the treatment, remission, and cure of patients. In this context, knowledge of epigenetic phenomena, their relationship with tumor cells and how the immune system can be epigenetically modulated represent some of the greatest advances in the development of anticancer therapies. Epigenetics is a rapidly growing field that studies how environmental factors can affect gene expression without altering DNA sequence. Epigenomic changes include DNA methylation, histone modifications, and non-coding RNA regulation, which impact cellular function. Epigenetics has shown promise in developing cancer therapies, such as immunotherapy, which aims to stimulate the immune system to attack cancer cells. For example, PD-1 and PD-L1 are biomarkers that regulate the immune response to cancer cells and recent studies have shown that epigenetic modifications can affect their expression, potentially influencing the efficacy of immunotherapy. New therapies targeting epigenetic modifications, such as histone deacetylases and DNA methyltransferases, are being developed for cancer treatment, and some have shown promise in preclinical studies and clinical trials. With growing understanding of epigenetic regulation, we can expect more personalized and effective cancer immunotherapies in the future. This review highlights key advances in the use of epigenetic and epigenomic tools and modern immuno-oncology strategies to treat several types of tumors.
Collapse
Affiliation(s)
| | | | - Michel Lopes Leite
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasília, DF, Brazil
- Department of Cell Biology, Institute of Biological Sciences, Campus Darcy Ribeiro, University of Brasilia (UnB), Brasília, DF, Brazil
| | - Nicolau B da Cunha
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasília, DF, Brazil
- Faculty of Agronomy and Veterinary Medicine (FAV), Campus Darcy Ribeiro, University of Brasilia (UnB), Brasília, DF, Brazil
- Graduate Program in Agronomy, Campus Darcy Ribeiro, University of Brasilia (UnB), Brasília, DF, Brazil
| | - Fabricio F Costa
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasília, DF, Brazil
- Cancer Biology and Epigenomics Program, Northwestern University’s Feinberg School of Medicine, Chicago, IL, USA
- Genomic Enterprise, San FranciscoCA, USA
| |
Collapse
|
366
|
Joseph GJ, Johnson DB, Johnson RW. Immune checkpoint inhibitors in bone metastasis: Clinical challenges, toxicities, and mechanisms. J Bone Oncol 2023; 43:100505. [PMID: 37842554 PMCID: PMC10568292 DOI: 10.1016/j.jbo.2023.100505] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the field of anti-cancer therapy over the last decade; they provide durable clinical responses against tumors by inhibiting immune checkpoint proteins that canonically regulate the T cell-mediated immune response. Despite their success in many primary tumors and soft tissue metastases, ICIs function poorly in patients with bone metastases, and these patients do not have the same survival benefit as patients with the same primary tumor type (e.g., non-small cell lung cancer [NSCLC], urothelial, renal cell carcinoma [RCC], etc.) that has not metastasized to the bone. Additionally, immune-related adverse events including rheumatologic and musculoskeletal toxicities, bone loss, and increased fracture risk develop after treatment with ICIs. There are few preclinical studies that investigate the interplay of the immune system in bone metastases; however, the current literature suggests a role for CD8+ T cells and myeloid cell subsets in bone homeostasis. As such, this review focuses on findings from the clinical and pre-clinical studies that have investigated immune checkpoint blockade in the bone metastatic setting and highlights the need for more comprehensive investigations into the relationship between immune cell subsets, ICIs, and the bone-tumor microenvironment.
Collapse
Affiliation(s)
- Gwenyth J. Joseph
- Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Douglas B. Johnson
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachelle W. Johnson
- Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
367
|
Backlund E, Grozman V, Egyhazi Brage S, Lewensohn R, Lindberg K, Helgadottir H. Radiotherapy with or without immunotherapy in metastatic melanoma: efficacy and tolerability. Acta Oncol 2023; 62:1921-1930. [PMID: 37966921 DOI: 10.1080/0284186x.2023.2280766] [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: 09/01/2023] [Accepted: 11/03/2023] [Indexed: 11/17/2023]
Abstract
INTRODUCTION Radiotherapy (RT) is primarily considered as a palliative treatment in patients with metastatic melanoma. However, observations suggest that when RT is combined with immune checkpoint inhibitors (ICI), it can induce an immune response leading to an anti-tumoral effect also distant from the irradiated area - a phenomenon called 'abscopal effect'. The frequency and circumstances of abscopal effect among metastatic melanoma patients remains uncertain and further research is necessary. MATERIAL AND METHOD This retrospective study included all metastatic melanoma patients who received non-stereotactic RT in Stockholm, Sweden in 2015-2020. Patients were grouped depending on if RT was given at start of ICI (RT + ICI(start)), at ICI progression (RT + ICI(salvage)) or without ICI (RT(only)). Response rates in irradiated (RR(irradiated)) and overall response rates in non-irradiated (ORR(non-irradiated)) metastases were evaluated together with survival and toxicity in each cohort. RESULTS In the RT + ICI(start) (n = 47), RT + ICI(salvage) (n = 41) and RT(only) (n = 55) cohorts, RR(irradiated) was 70.7%, 67.5% and 43.1% (p = 0.018) while the ORR(non-irradiated) was 36.1%, 14.8% and 0.0% (p = 0.003), and the median overall survival was 18.2, 15.0 and 7.2 months, respectively (p = 0.014). Local response to RT was in all cohorts associated with longer survival (p < 0.001). The frequency of grade ≥3 immune-related adverse events was 17.0% and 19.5% in the RT + ICI(start) and RT + ICI(salvage) cohorts. No increased frequency of RT-related adverse events was seen in the RT + ICI cohorts, compared to the RT(only) cohort. CONCLUSION This retrospective study showed that melanoma patients receiving RT in combination with ICI had a superior antitumoral response in both irradiated and non-irradiated lesions as compared to patients receiving only RT. Additionally, a subgroup of patients receiving RT when progressing on ICI experienced tumor regression also in non-irradiated areas.
Collapse
Affiliation(s)
- Ellen Backlund
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Vitali Grozman
- Department of Diagnostic Radiology, Karolinska University Hospital, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm
| | | | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Lindberg
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hildur Helgadottir
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
368
|
Yang L, Atakhanova N, Arellano MTC, Mohamed MY, Hani T, Fahdil AA, Castillo-Acobo RY, Juyal A, Hussein AK, Amin AH, Pecho RDC, Akhavan-Sigari R. Translational research of new developments in targeted therapy of colorectal cancer. Pathol Res Pract 2023; 252:154888. [PMID: 37948996 DOI: 10.1016/j.prp.2023.154888] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
A severe global health concern is the rising incidence and mortality rate of colorectal cancer (CRC). Chemotherapy, which is typically used to treat CRC, is known to have limited specificity and can have noticeable side effects. A paradigm shift in cancer treatment has been brought about by the development of targeted therapies, which has led to the appearance of pharmacological agents with improved efficacy and decreased toxicity. Epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), and BRAF are among the molecular targets covered in this review that are used in targeted therapy for CRC. The current discussion also covers advancements in targeted therapeutic approaches, such as antibody-drug conjugates, immune checkpoint inhibitors, and chimeric antigen receptor (CAR) T-cell therapy. A review of the clinical trials and application of these particular therapies in treating CRC is also done. Despite the improvements in targeted therapy for CRC, problems such as drug resistance and patient selection remain to be solved. Despite this, targeted therapies have offered fresh possibilities for identifying and treating CRC, paving the way for the development of personalized medicine and extending the life expectancy and general well-being of CRC patients.
Collapse
Affiliation(s)
- Lei Yang
- Department of Clinical Laboratory, People's Hospital of Chongqing Liangjiang New Area, Chongqing 401121, China
| | - Nigora Atakhanova
- Head of the Department of Oncology, Tashkent Medical Academy, Tashkent 100109, Uzbekistan
| | | | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Iraq
| | | | - Ashima Juyal
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland
| |
Collapse
|
369
|
Mollica V, Rizzo A, Marchetti A, Tateo V, Tassinari E, Rosellini M, Massafra R, Santoni M, Massari F. The impact of ECOG performance status on efficacy of immunotherapy and immune-based combinations in cancer patients: the MOUSEION-06 study. Clin Exp Med 2023; 23:5039-5049. [PMID: 37535194 DOI: 10.1007/s10238-023-01159-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
ECOG performance status (PS) is a pivotal prognostic factor in a wide number of solid tumors. We performed a meta-analysis to assess the role of ECOG PS in terms of survival in patients with ECOG PS 0 or ECOG PS 1 treated with immunotherapy alone or combined with other anticancer treatments. Following the Preferred Reporting Items for Systematic Reviews and Meta-analyses, all phase II and III randomized clinical trials that compared immunotherapy or immune-based combinations in patients with solid tumors were retrieved. The outcomes of interest were overall survival (OS) and progression-free survival (PFS). We also performed subgroup analyses focused on type of therapy (ICI monotherapy or combinations), primary tumor type, setting (first line of treatment, subsequent lines). Overall, 60 studies were included in the analysis for a total of 35.020 patients. The pooled results showed that immunotherapy, either alone or in combination, reduces the risk of death or progression in both ECOG PS 0 and 1 populations. The survival benefit was consistent in all subgroups. Immune checkpoint inhibitors monotherapy or immune-based combinations are associated with improved survival irrespective of ECOG PS 0 or 1. Clinical trials should include more frail patients to assess the value of immunotherapy in these patients.
Collapse
Affiliation(s)
- Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Valentina Tateo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | | | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
370
|
Sada I, Harada Y, Hiyama T, Mizukami M, Kan T, Kawai M, Kiuchi Y. Uveitis associated with immune checkpoint inhibitors or BRAF/MEK inhibitors in patients with malignant melanoma. Melanoma Res 2023; 33:539-546. [PMID: 37788106 DOI: 10.1097/cmr.0000000000000933] [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: 10/05/2023]
Abstract
The objective of this study was to evaluate the frequency and characteristics of uveitis associated with immune checkpoint inhibitors (ICIs) or BRAF/MEK inhibitors (B/MIs) in patients with malignant melanoma. Patients diagnosed with malignant melanoma who underwent radical or local resection for malignant melanoma, regardless of clinical stage or postoperative adjuvant therapy, at Hiroshima University Hospital from January 2015 to June 2021 were enrolled in a retrospective cohort. The medical records of patients were collected to estimate the prevalence of ocular adverse events. The clinical characteristics of patients who developed uveitis were reviewed. Among 152 patients, 54 and 12 were treated with ICIs and B/MIs, respectively. Four patients developed uveitis; 1 in the ICI group and 3 in the B/MI group, while there were no uveitis cases among patients who did not receive ICIs or B/MIs. Three patients had Vogt-Koyanagi-Harada disease-like findings. Uveitis was improved by steroid therapy with or without oncological treatment interruption. Oncological treatment could be resumed. Patients with melanoma treated with ICIs or B/MIs had a higher risk of uveitis compared with those who did not receive them. Oncological treatment could be resumed in all patients who developed uveitis.
Collapse
Affiliation(s)
- Ikuyo Sada
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University
| | - Yosuke Harada
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University
| | - Tomona Hiyama
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University
| | - Mina Mizukami
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University
| | - Takanobu Kan
- Department of Dermatology, Graduate School of Biomedical Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Mikio Kawai
- Department of Dermatology, Graduate School of Biomedical Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology and Visual Science, Graduate School of Biomedical Sciences, Hiroshima University
| |
Collapse
|
371
|
Badrigilan S, Meola A, Chang SD, Rezaeian S, Nemati H, Almasi T, Rostampour N. Stereotactic radiosurgery with immune checkpoint inhibitors for brain metastases: a meta-analysis study. Br J Neurosurg 2023; 37:1533-1543. [PMID: 34979828 DOI: 10.1080/02688697.2021.2022098] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/20/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are an emerging tool in the treatment of brain metastases (BMs), Stereotactic radiosurgery (SRS), traditionally used for BMs, elicits an immune brain response and can act synergistically with ICIs. We aim to investigate the efficacy of ICI administered with SRS and determine the impact of timing on BM response. METHODS A systematical search was performed to identify potential studies concerning BMs managed with SRS alone or with SRS + ICI with relative timing administration (ICI concurrent with SRS, ICI nonconcurrent with SRS, SRS before ICI, SRS after ICI). The overall survival (OS), 12-month OS, local progression-free survival (LPFS), 12-month local brain control (LBC), distant progression-free survival (DPFS), 12-month distant brain control (DBC), and adverse events (intracranial hemorrhage, radionecrosis) were analyzed using the random-effects model. RESULTS A total of 16 retrospective studies with 1356 BM patients were included. Compared to nonconcurrent therapy, concurrent therapy revealed a significantly longer OS (HR= 1.43; p = 0.008) and 12-months LBC (HR = 1.91; p = 0.04), a similar 12-months DBC (HR = 1.12; p = 0.547) and higher complication rate (R = 0.77; p = 0.346). Concurrent therapy leads to a significantly higher OS compared to ICI before SRS (HR = 2.55; p = 0.0003). CONCLUSION The combination of SRS with ICI improves patients' clinical and radiological outcomes. The effectiveness of the combination is subject to the identification of an optimal therapeutic window.
Collapse
Affiliation(s)
- Samireh Badrigilan
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Antonio Meola
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven D Chang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Shahab Rezaeian
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Nemati
- Department of Epidemiology, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Tinoosh Almasi
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nima Rostampour
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
372
|
Muto I, Koga H, Doi R, Katayama E, Nakama K, Nakama T. Efficacy of Nivolumab and Ipilimumab Combined Therapy as a First-Line Therapy for Patients with Advanced Melanoma and the Urgent Need for an Effective Second-Line Therapy for Patients with Wild-Type BRAF in Japan: A Single Center Retrospective Study. Kurume Med J 2023; 69:75-80. [PMID: 37793888 DOI: 10.2739/kurumemedj.ms6912008] [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: 10/06/2023]
Abstract
Therapeutic advantages of immune checkpoint inhibitors, anti-programmed death-1 (PD-1), and anticytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) in melanoma have been reported recently. In this study, we conducted a retrospective study to evaluate the clinical efficacy and safety of the combined use of nivolumab and ipilimumab as a first-line therapy for Japanese patients with advanced melanoma. Moreover, we examined the effects of second-line treatment. Seven patients were enrolled in this study. The median progression-free survival (PFS) and median overall survival (OS) were 7 months (95%CI, 1.868-12.132) and 12 months (95%CI, 0.000- 27.397), respectively. The objective response rate (ORR) and the disease control rate (DCR) were 42.9 % and 85.7 %. Three patients chose pembrolizumab monotherapy as second-line therapy after the combination therapy due to their BRAF wild-type status, which resulted in progressive disease. ORR and DCR were 0% and 33.3%, respectively, with pembrolizumab. Grade 3 or 4 immune-related adverse events occurred in 71.4% of the patients treated with the combined-therapy. All irAEs were treated with corticosteroid or hormone replacement therapy. Although this single center retrospective study had some limitations, it demonstrated good efficacy for the combined use of nivolumab and ipilimumab as a first-line therapy for Japanese patients with advanced melanoma. Moreover, poor efficacy was observed for the second-line therapy after the combined therapy. These findings suggest that a novel second-line therapy is required for patients with advanced melanoma in Japan, particularly for patients with wildtype BRAF.
Collapse
Affiliation(s)
- Ikko Muto
- Department of Dermatology, Kurume University School of Medicine
| | - Hiroshi Koga
- Department of Dermatology, Kurume University School of Medicine
| | - Reiichi Doi
- Department of Dermatology, Kurume University School of Medicine
| | - Eri Katayama
- Department of Dermatology, Kurume University School of Medicine
| | - Kenta Nakama
- Department of Dermatology, Kurume University School of Medicine
| | - Takekuni Nakama
- Department of Dermatology, Kurume University School of Medicine
| |
Collapse
|
373
|
Morimoto K, Yamada T, Takayama K. The landscape of immune therapy in vulnerable patients with advanced non-small cell lung cancer: a narrative review. Transl Lung Cancer Res 2023; 12:2310-2321. [PMID: 38090528 PMCID: PMC10713257 DOI: 10.21037/tlcr-23-581] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/07/2023] [Indexed: 08/27/2024]
Abstract
BACKGROUND AND OBJECTIVE The clinical development of immune checkpoint inhibitors (ICIs) has led to substantial advances in the treatment of lung cancer. In particular, the contribution of ICIs to the long-term survival of certain patients with non-small cell lung cancer (NSCLC) has been reported. With the accumulated experience in the use of ICIs, numerous studies have documented the efficacy and safety of ICIs in patients with diverse backgrounds, including those with problematic indications for drug therapy. In the current review, we summarize the most recent literature-based findings on ICI administration in vulnerable patients with NSCLC and provide an overview of the current status and prospects of ICIs. METHODS Herein, we defined vulnerable as the group of patients with NSCLC and performance status (PS) ≥2 (poor PS), advanced age (≥75 years), or cancer cachexia. We conducted a narrative review of the literature on the efficacy and safety of ICIs in vulnerable patients with advanced NSCLC. KEY CONTENT AND FINDINGS Among the vulnerable patient group, poor PS was a strong, poor prognostic factor, even in patients undergoing ICI therapy. ICI therapy in older patients can be effective, although adverse events (AEs) should be carefully monitored. The efficacy of ICI therapy in patients with cancer cachexia is poor, with further therapeutic development warranted. CONCLUSIONS Although prior studies have evaluated lung cancer pharmacotherapy in various vulnerable populations, clinical studies on the application of ICIs in patients with vulnerable NSCLC are lacking in both number and quality. Further development of these therapeutic agents, with the emergence of additional evidence regarding their appropriate use in this population, is expected.
Collapse
Affiliation(s)
- Kenji Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
374
|
Zhou Y, Ding S. Key Determinants of Immune-Mediated Adverse Reactions to Oncology Drugs. Cancers (Basel) 2023; 15:5622. [PMID: 38067327 PMCID: PMC10705334 DOI: 10.3390/cancers15235622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/23/2023] [Accepted: 11/23/2023] [Indexed: 06/16/2024] Open
Abstract
To overcome the epidemiological severity of cancer, developing effective treatments is urgently required. In response, immune checkpoint inhibitors (ICIs) have been revealed as a promising resolution for treatment-resistant cancers across the world. Yet, they have both advantages and disadvantages, bringing therapeutic benefits while simultaneously inducing toxicity, and in particular, immune-mediated adverse drug reactions (imADRs), to the human body. These imADRs can be pathogenic and sometimes lethal, hampering health prediction and monitoring following the provision of ICI treatment. Therefore, it is necessary to collectively identify the determinant factors that contribute to these imADRs induced by ICIs. This article evaluated treatment-, tumor-, and patient-related determinants, and indicated a research gap for future investigations on the pathogenic mechanism of imADRs and translational conversion of determinants into clinical biomarkers to aid pharmacovigilance and cancer therapies.
Collapse
Affiliation(s)
- Yihan Zhou
- Medical Sciences Division, Department of Oncology, University of Oxford, Old Road Campus Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Shan Ding
- Department of Life Science, Imperial College London, South Kensington Campus, Exhibition Road, London SW7 2AZ, UK;
| |
Collapse
|
375
|
Tsakmaklis A, Farowski F, Zenner R, Lesker TR, Strowig T, Schlößer H, Lehmann J, von Bergwelt-Baildon M, Mauch C, Schlaak M, Knuever J, Schweinsberg V, Heinzerling LM, Vehreschild MJGT. TIGIT + NK cells in combination with specific gut microbiota features predict response to checkpoint inhibitor therapy in melanoma patients. BMC Cancer 2023; 23:1160. [PMID: 38017389 PMCID: PMC10685659 DOI: 10.1186/s12885-023-11551-5] [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: 07/29/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Composition of the intestinal microbiota has been correlated to therapeutic efficacy of immune checkpoint inhibitors (ICI) in various cancer entities including melanoma. Prediction of the outcome of such therapy, however, is still unavailable. This prospective, non-interventional study was conducted in order to achieve an integrated assessment of the connection between a specific intestinal microbiota profile and antitumor immune response to immune checkpoint inhibitor therapy (anti-PD-1 and/or anti-CTLA-4) in melanoma patients. METHODS We assessed blood and stool samples of 29 cutaneous melanoma patients who received immune checkpoint inhibitor therapy. For functional and phenotypical immune analysis, 12-color flow cytometry and FluoroSpot assays were conducted. Gut microbiome was analyzed with shotgun metagenomics sequencing. To combine clinical, microbiome and immune variables, we applied the Random Forest algorithm. RESULTS A total of 29 patients was analyzed in this study, among whom 51.7% (n = 15) reached a durable clinical benefit. The Immune receptor TIGIT is significantly upregulated in T cells (p = 0.0139) and CD56high NK cells (p = 0.0037) of responders. Several bacterial taxa were associated with response (e.g. Ruminococcus torques) or failure (e.g. Barnesiella intestinihominis) to immune therapy. A combination of two microbiome features (Barnesiella intestinihominis and the Enterobacteriaceae family) and one immune feature (TIGIT+ CD56high NK cells) was able to predict response to ICI already at baseline (AUC = 0.85; 95% CI: 0.841-0.853). CONCLUSIONS Our results reconfirm a link between intestinal microbiota and response to ICI therapy in melanoma patients and furthermore point to TIGIT as a promising target for future immunotherapies.
Collapse
Affiliation(s)
- Anastasia Tsakmaklis
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany
| | - Fedja Farowski
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt, 60590, Germany
| | - Rafael Zenner
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Till Robin Lesker
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Strowig
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Hans Schlößer
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Interventional Immunology, University of Cologne, Cologne, Germany
| | - Jonas Lehmann
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Interventional Immunology, University of Cologne, Cologne, Germany
| | | | - Cornelia Mauch
- Department of Dermatology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Max Schlaak
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Jana Knuever
- Department of Dermatology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Viola Schweinsberg
- Department of Dermatology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lucie M Heinzerling
- Department of Dermatology and Allergy, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Maria J G T Vehreschild
- Department I of Internal Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany.
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt, 60590, Germany.
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine & Pharmacology ITMP, Frankfurt am Main, 60596, Germany.
| |
Collapse
|
376
|
Niessner H, Hüsch A, Kosnopfel C, Meinhardt M, Westphal D, Meier F, Schilling B, Sinnberg T. Exploring the In Vitro and In Vivo Therapeutic Potential of BRAF and MEK Inhibitor Combination in NRAS-Mutated Melanoma. Cancers (Basel) 2023; 15:5521. [PMID: 38067230 PMCID: PMC10705743 DOI: 10.3390/cancers15235521] [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: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 10/16/2024] Open
Abstract
INTRODUCTION Patients with NRAS-mutant metastatic melanoma often have an aggressive disease requiring a fast-acting, effective therapy. The MEK inhibitor binimetinib shows an overall response rate of 15% in patients with NRAS-mutant melanoma, providing a backbone for combination strategies. Our previous studies demonstrated that in NRAS-mutant melanoma, the antitumor activity of the MEK inhibitor binimetinib was significantly potentiated by the BRAFV600E/K inhibitor encorafenib through the induction of ER stress, leading to melanoma cell death by apoptotic mechanisms. Encorafenib combined with binimetinib was well tolerated in a phase III trial showing potent antitumor activity in BRAF-mutant melanoma, making a rapid evaluation in NRAS-mutant melanoma imminently feasible. These data provide a mechanistic rationale for the evaluation of binimetinib combined with encorafenib in preclinical and clinical studies on NRAS-mutant metastatic melanoma. METHODS The combination of BRAFi plus MEKi was tested in a monolayer culture of patient-derived cell lines and in corresponding patient-derived tissue slice cultures of NRAS-mutant melanoma. To investigate the treatment in vivo, NSG (NOD. Cg-PrkdcscidIl2rgtm1Wjl/SzJ) mice were subcutaneously injected with three different BRAF wild-type melanoma models harboring oncogenic NRAS mutations and treated orally with encorafenib (6 mg/kg body weight, daily) with or without binimetinib (8 mg/kg body weight, twice daily). In parallel, an individual healing attempt was carried out by treating one patient with an NRAS-mutated tumor. RESULTS Encorafenib was able to enhance the inhibitory effect on cell growth of binimetinib only in the cell line SKMel147 in vitro. It failed to enhance the apoptotic effect found in two other NRAS-mutated cell lines. Encorafenib led to a hyperactivation of ERK which could be reduced with the combinational treatment. In two of the three patient-derived tissue slice culture models of NRAS-mutant melanomas, a slight tendency of a combinatorial effect was seen which was not significant. Encorafenib showed a slight induction of the ER stress genes ATF4, CHOP, and NUPR1. The combinational treatment was able to enhance this effect, but not significantly. In the mouse model, the combination therapy of encorafenib with binimetinib resulted in reduced tumor growth compared to the control and encorafenib groups; however, the best effect in terms of tumor growth inhibition was measured in the binimetinib therapy group. The therapy showed no effect in an individual healing attempt for a patient suffering from metastatic, therapy-refractory NRAS-mutated melanoma. CONCLUSION In in vitro and ex vivo settings, the combination therapy was observed to elicit a response; however, it did not amplify the efficacy observed with binimetinib alone, whereas in a patient, the combinational treatment remained ineffective. The preclinical in vivo data showed no increased combinatorial effect. However, the in vivo effect of binimetinib as monotherapy was unexpectedly high in the tested regimen. Nevertheless, binimetinib proved to be advantageous in the treatment of melanoma in vivo and led to high rates of apoptosis in vitro; hence, it still seems to be a good base for combination with other substances in the treatment of patients with NRAS-mutant melanoma.
Collapse
Affiliation(s)
- Heike Niessner
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
| | - Anna Hüsch
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
| | - Corinna Kosnopfel
- Department of Hematology, Oncology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Matthias Meinhardt
- Department of Pathology, Medical Faculty and University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany;
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
| | - Dana Westphal
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (F.M.)
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, 01307 Dresden, Germany
- Center for Regenerative Therapies Dresden, TU Dresden, 01307 Dresden, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany;
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
377
|
Li H, Zhu Y, Wang X, Feng Y, Qian Y, Ma Q, Li X, Chen Y, Chen K. Joining Forces: The Combined Application of Therapeutic Viruses and Nanomaterials in Cancer Therapy. Molecules 2023; 28:7679. [PMID: 38005401 PMCID: PMC10674375 DOI: 10.3390/molecules28227679] [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: 09/29/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer, on a global scale, presents a monumental challenge to our healthcare systems, posing a significant threat to human health. Despite the considerable progress we have made in the diagnosis and treatment of cancer, realizing precision cancer therapy, reducing side effects, and enhancing efficacy remain daunting tasks. Fortunately, the emergence of therapeutic viruses and nanomaterials provides new possibilities for tackling these issues. Therapeutic viruses possess the ability to accurately locate and attack tumor cells, while nanomaterials serve as efficient drug carriers, delivering medication precisely to tumor tissues. The synergy of these two elements has led to a novel approach to cancer treatment-the combination of therapeutic viruses and nanomaterials. This advantageous combination has overcome the limitations associated with the side effects of oncolytic viruses and the insufficient tumoricidal capacity of nanomedicines, enabling the oncolytic viruses to more effectively breach the tumor's immune barrier. It focuses on the lesion site and even allows for real-time monitoring of the distribution of therapeutic viruses and drug release, achieving a synergistic effect. This article comprehensively explores the application of therapeutic viruses and nanomaterials in tumor treatment, dissecting their working mechanisms, and integrating the latest scientific advancements to predict future development trends. This approach, which combines viral therapy with the application of nanomaterials, represents an innovative and more effective treatment strategy, offering new perspectives in the field of tumor therapy.
Collapse
Affiliation(s)
- Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
- Ocean College, Beibu Gulf University, Qinzhou 535011, China
| | - Yunhuan Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xin Wang
- Center of Infectious Disease Research, School of Life Science, Westlake University, Hangzhou 310024, China;
| | - Yilu Feng
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yuncheng Qian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Qiman Ma
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xinyuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yihan Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| |
Collapse
|
378
|
Chai F, Li P, He Y, Zhou Z, Guo S, Liu X, Zhou L, Ren H. Genetically incorporated crosslinkers identify regulators of membrane protein PD-L1 in mammalian cells. Cell Chem Biol 2023; 30:1488-1497.e5. [PMID: 37541256 DOI: 10.1016/j.chembiol.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/01/2023] [Accepted: 07/16/2023] [Indexed: 08/06/2023]
Abstract
Profiling membrane proteins' interacting networks is crucial for understanding their regulatory mechanisms and functional characteristics, but it remains a challenging task. Here, by combining genetic incorporation of crosslinkers, tandem denatured purification, and proteomics, we added interaction partners for PD-L1, a cancer cell surface protein that inhibits T cell activity. The site-specifically incorporated crosslinker mediates the covalent capture of interactions under physiological conditions and enabled the PD-L1 complexes to withstand the harsh extraction conditions of membrane proteins. Subsequent experiments led to the identification of potential PD-L1 interaction candidates and verified membrane-associated progesterone receptor component 1 as a novel PD-L1 interaction partner in mammalian cells. Importantly, we demonstrated that PGRMC1 positively regulates PD-L1 expression by regulating GSK3β-mediated PD-L1 degradation in cancer cells. Furthermore, PGRMC1 knockdown results in dramatically enhanced T cell-mediated cytotoxicity in cancer cells. In conclusion, our study elucidated the interactome of PD-L1 and uncovered a new player in the PD-L1 regulation mechanism.
Collapse
Affiliation(s)
- Fangni Chai
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Pan Li
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yong He
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhihui Zhou
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Shupan Guo
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xin Liu
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Li Zhou
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Haiyan Ren
- Division of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China; Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
379
|
Lim KK, Ng K, Balachandran S, Russell MD, Boalch A, Sinclair A, Coker B, Vinnakota K, Mansoor R, Douiri A, Marks LV, Sacks S. Measuring the impact of monoclonal antibody therapies. Front Med (Lausanne) 2023; 10:1256712. [PMID: 38046416 PMCID: PMC10690588 DOI: 10.3389/fmed.2023.1256712] [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: 07/11/2023] [Accepted: 10/13/2023] [Indexed: 12/05/2023] Open
Abstract
Objective Monoclonal antibody (Mab) treatments have significantly improved the quality and quantity of life, but they are some of the most expensive treatments, resulting in a degree of hesitancy to introduce new Mab agents. A system for estimating the effect of Mab drugs, in general, would optimally inform health strategy and fully realize how a single scientific discovery can deliver health benefits. We evaluated such a method with several well-established Mab regimens. Methods We selected five different Mab regimens in oncology and rheumatology in England. We carried out two systematic literature reviews and meta-analyses to assess health outcomes (Health Assessment Questionnaire-Disability Index for rheumatoid arthritis; overall mortality for melanoma) from real-world data and compared them to the outcomes from randomized control trials (RCTs). We applied economic modeling to estimate the net monetary benefits for health outcomes for the estimated patient population size for each Mab regimen. Results Meta-analyses of 27 eligible real-world data (RWD) sets and 26 randomized controlled trial (RCT) sets found close agreement between the observed and expected health outcomes. A Markov model showed the net positive monetary benefit in three Mab regimens and the negative benefit in two regimens. However, because of limited access to NHS data, the economic model made several assumptions about the number of treated patients and the cost of treatment to the NHS, the accuracy of which may affect the estimation of the net monetary benefit. Conclusion RCT results reliably inform the real-world experience of Mab treatments. Calculation of the net monetary benefit by the algorithm described provides a valuable overall measure of the health impact, subject to the accuracy of data inputs. This study provides a compelling case for building a comprehensive, systematized, and accessible database and related analytics, on all Mab treatments within health services.
Collapse
Affiliation(s)
- Ka Keat Lim
- Department of Population Health Sciences, King's College London, London, United Kingdom
| | - Kenrick Ng
- Cancer Institute, University College London, London, United Kingdom
| | | | - Mark D. Russell
- Centre for Rheumatic Diseases, King's College London, London, United Kingdom
| | - Amy Boalch
- Centre for Rheumatic Diseases, King's College London, London, United Kingdom
| | - Alasdair Sinclair
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Bolaji Coker
- NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Krishna Vinnakota
- University College London Hospitals NHS Foundation Trust (UCLH), London, United Kingdom
| | - Rashid Mansoor
- Department of Population Health Sciences, King's College London, London, United Kingdom
| | - Abdel Douiri
- Department of Population Health Sciences, King's College London, London, United Kingdom
| | | | - Steven Sacks
- Peter Gorer Department of Immunology, King's College London, London, United Kingdom
| |
Collapse
|
380
|
In GK, Ribeiro JR, Yin J, Xiu J, Bustos MA, Ito F, Chow F, Zada G, Hwang L, Salama AKS, Park SJ, Moser JC, Darabi S, Domingo-Musibay E, Ascierto ML, Margolin K, Lutzky J, Gibney GT, Atkins MB, Izar B, Hoon DSB, VanderWalde AM. Multi-omic profiling reveals discrepant immunogenic properties and a unique tumor microenvironment among melanoma brain metastases. NPJ Precis Oncol 2023; 7:120. [PMID: 37964004 PMCID: PMC10646102 DOI: 10.1038/s41698-023-00471-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023] Open
Abstract
Melanoma brain metastases (MBM) are clinically challenging to treat and exhibit variable responses to immune checkpoint therapies. Prior research suggests that MBM exhibit poor tumor immune responses and are enriched in oxidative phosphorylation. Here, we report results from a multi-omic analysis of a large, real-world melanoma cohort. MBM exhibited lower interferon-gamma (IFNγ) scores and T cell-inflamed scores compared to primary cutaneous melanoma (PCM) or extracranial metastases (ECM), which was independent of tumor mutational burden. Among MBM, there were fewer computationally inferred immune cell infiltrates, which correlated with lower TNF and IL12B mRNA levels. Ingenuity pathway analysis (IPA) revealed suppression of inflammatory responses and dendritic cell maturation pathways. MBM also demonstrated a higher frequency of pathogenic PTEN mutations and angiogenic signaling. Oxidative phosphorylation (OXPHOS) was enriched in MBM and negatively correlated with NK cell and B cell-associated transcriptomic signatures. Modulating metabolic or angiogenic pathways in MBM may improve responses to immunotherapy in this difficult-to-treat patient subset.
Collapse
Affiliation(s)
- Gino K In
- Division of Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | | | - Jun Yin
- Caris Life Sciences, Phoenix, AZ, USA
| | | | - Matias A Bustos
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Fumito Ito
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frances Chow
- Department of Neurology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurological Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gabriel Zada
- Department of Neurological Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lindsay Hwang
- LAC+USC Medical Center, Los Angeles, CA, USA
- Department of Radiation Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - April K S Salama
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Soo J Park
- Division of Hematology/Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Justin C Moser
- HonorHealth Research and Innovation Institute, Scottsdale, AZ, USA
| | - Sourat Darabi
- Hoag Family Cancer Institute, Hoag Hospital, Newport Beach, CA, USA
| | - Evidio Domingo-Musibay
- Department of Medicine, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Maria L Ascierto
- Rosalie and Harold Rae Brown Cancer Immunotherapy Research Program, Borstein Family Melanoma Program, Department of Translational Immunology, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Kim Margolin
- Department of Medical Oncology, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Jose Lutzky
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | - Geoffrey T Gibney
- Division of Hematology and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC, USA
| | - Michael B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Benjamin Izar
- Columbia University, Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Ari M VanderWalde
- Caris Life Sciences, Irving, TX, USA
- West Cancer Center and Research Institute, 514 Chickasawba St., Blytheville, Arkansas, 72315, USA
| |
Collapse
|
381
|
Bodensohn R, Maier SH, Belka C, Minniti G, Niyazi M. Stereotactic Radiosurgery of Multiple Brain Metastases: A Review of Treatment Techniques. Cancers (Basel) 2023; 15:5404. [PMID: 38001664 PMCID: PMC10670108 DOI: 10.3390/cancers15225404] [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: 08/31/2023] [Revised: 10/22/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
The advancement of systemic targeted treatments has led to improvements in the management of metastatic disease, particularly in terms of survival outcomes. However, brain metastases remain less responsive to systemic therapies, underscoring the significance of local interventions for comprehensive disease control. Over the past years, the threshold for treating brain metastases through stereotactic radiosurgery has risen. Yet, as the number of treated metastases increases, treatment complexity and duration also escalate. This trend has made multi-isocenter radiosurgery treatments, such as those with the Gamma Knife, challenging to plan and lengthy for patients. In contrast, single-isocenter approaches employing linear accelerators offer an efficient and expeditious treatment option. This review delves into the literature, comparing different linear-accelerator-based techniques with each other and in relation to dedicated systems, focusing on dosimetric considerations and feasibility.
Collapse
Affiliation(s)
- Raphael Bodensohn
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany;
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Sebastian H. Maier
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (S.H.M.); (C.B.)
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (S.H.M.); (C.B.)
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership between DKFZ and LMU University Hospital, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Giuseppe Minniti
- IRCCS Neuromed, 86077 Pozzilli, Italy;
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany;
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, 72076 Tübingen, Germany
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (S.H.M.); (C.B.)
- German Cancer Consortium (DKTK), Partner Site Tübingen, A Partnership between DKFZ and University Hospital, 72076 Tübingen, Germany
| |
Collapse
|
382
|
Huang X, Hu M, Sun T, Li J, Zhou Y, Yan Y, Xuan B, Wang J, Xiong H, Ji L, Zhu X, Tong T, Ning L, Ma Y, Zhao Y, Ding J, Guo Z, Zhang Y, Fang JY, Hong J, Chen H. Multi-kingdom gut microbiota analyses define bacterial-fungal interplay and microbial markers of pan-cancer immunotherapy across cohorts. Cell Host Microbe 2023; 31:1930-1943.e4. [PMID: 37944495 DOI: 10.1016/j.chom.2023.10.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/11/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
The effect of gut bacteria on the response to immune checkpoint inhibitors (ICIs) has been studied, but the relationship between fungi and ICI responses is not fully understood. Herein, 862 fecal metagenomes from 9 different cohorts were integrated for the identification of differentially abundant fungi and subsequent construction of random forest (RF) models to predict ICI responses. Fungal markers demonstrate excellent performance, with an average area under the curve (AUC) of 0.87. Their performance improves even further, reaching an average AUC of 0.89 when combined with bacterial markers. Higher enrichment of exhausted T cells is detected in responders, as predicted by fungal markers. Multi-kingdom network and functional analysis reveal that the fungus Schizosaccharomyces octosporus may ferment starch into short-chain fatty acids in responders. This study provides a fungal profile of the ICI response and the identification of multi-kingdom microbial markers with good performance that may improve the overall applicability of ICI therapy.
Collapse
Affiliation(s)
- Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Tiantian Sun
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jiantao Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yilu Zhou
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yuqing Yan
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jilin Wang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Hua Xiong
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Linhua Ji
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China.
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Shanghai, China.
| |
Collapse
|
383
|
Li B, Wang S, Shan B, Li B, Li F. A PD-L1xCD3 bispecific nanobody as a novel T-cell engager in treating PD-L1 overexpression melanoma. Mol Immunol 2023; 163:20-27. [PMID: 37722180 DOI: 10.1016/j.molimm.2023.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/11/2023] [Accepted: 09/07/2023] [Indexed: 09/20/2023]
Abstract
The development of Immune checkpoint blockade(ICB) therapy and BRAF- and MEK-targeted therapies has reshaped the survival outcomes of the patients with advanced melanoma. PD-1/PD-L1 blockade was an approved strategy in melanoma treatment. Here we design a PD-L1 xCD3 nanobody as a novel bispecific T cell engager (BiTE) in treating PD-L1 overexpression melanoma. BiTE PD-L1×CD3 Nb was predicted to bind near a large acidic surface on CD3-ε similar to UCHT1-scFv antibody based on alpha-fold and molecular docking. BiTE PD-L1×CD3 Nb and anti-CD3 Nb retained the ability to activate T cells to produce TNF-α and IFN-γ in a dose-dependent manner. The IC50 value of BiTE PD-L1×CD3 Nb was 4.208μg/mL. BiTE PD-L1×CD3 Nb showed obvious cytotoxic activity on both A375WT and A375PD-L1 related to PD-L1 expression level.
Collapse
Affiliation(s)
- Boping Li
- Department of Dermatology, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Shuang Wang
- Department of Dermatology, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Baihui Shan
- Department of Dermatology, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Baizhi Li
- Institute of Frontier Medical Science, School of Pharmaceutical Science, Jilin University, Changchun, China.
| | - Fuqiu Li
- Department of Dermatology, Second Affiliated Hospital of Jilin University, Changchun, China.
| |
Collapse
|
384
|
Huang C, Zhang K, Guo Y, Shen C, Liu X, Huang H, Dou X, Yu B. The crucial roles of m 6A RNA modifications in cutaneous cancers: Implications in pathogenesis, metastasis, drug resistance, and targeted therapies. Genes Dis 2023; 10:2320-2330. [PMID: 37554186 PMCID: PMC10404882 DOI: 10.1016/j.gendis.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/11/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022] Open
Abstract
N6-methyladenosine (m6A) is the most abundant internal modification on RNA. It is a dynamical and reversible process, which is regulated by m6A methyltransferase and m6A demethylase. The m6A modified RNA can be specifically recognized by the m6A reader, leading to RNA splicing, maturation, degradation or translation. The abnormality of m6A RNA modification is closely related to a variety of biological processes, especially the occurrence and development of tumors. Recent studies have shown that m6A RNA modification is involved in the pathogenesis of skin cancers. However, the precise molecular mechanisms of m6A-mediated cutaneous tumorigenesis have not been fully elucidated. Therefore, this review will summarize the biological characteristics of m6A modification, its regulatory role and mechanism in skin cancers, and the recent research progress of m6A-related molecular drugs, aiming to provide new ideas for clinical diagnosis and targeted therapy of cutaneous cancers.
Collapse
Affiliation(s)
- Cong Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Kaoyuan Zhang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Yang Guo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Changbing Shen
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Xiaoming Liu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Haiyan Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Xia Dou
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| |
Collapse
|
385
|
Kojima M. Variable selection using inverse probability of censoring weighting. Stat Methods Med Res 2023; 32:2184-2206. [PMID: 37675496 DOI: 10.1177/09622802231199335] [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: 09/08/2023]
Abstract
In this article, we propose two variable selection methods for adjusting the censoring information for survival times, such as the restricted mean survival time. To adjust for the influence of censoring, we consider an inverse probability of censoring weighted for subjects with events. We derive a least absolute shrinkage and selection operator (lasso)-type variable selection method, which considers an inverse weighting for of the squared losses, and an information criterion-type variable selection method, which applies an inverse weighting of the survival probability to the power of each density function in the likelihood function. We prove the consistency of the inverse probability of censoring weighted lasso estimator and the maximum inverse probability of censoring weighted likelihood estimator. The performance of the inverse probability of censoring weighted lasso and inverse probability of censoring weighted information criterion are evaluated via a simulation study with six scenarios, and then their variable selection ability is demonstrated using data from two clinical studies. The results confirm that inverse probability of censoring weighted lasso and the inverse probability of censoring weighted likelihood function produce good estimation accuracy and consistent variable selection. We conclude that our two proposed methods are useful variable selection tools for adjusting the censoring information for survival time analyses.
Collapse
Affiliation(s)
- Masahiro Kojima
- Biometrics Department, R&D Division, Kyowa Kirin Co. Ltd., Chiyoda-ku, Tokyo, Japan
- The Institute of Statistical Mathematics, Tachikawa, Tokyo, Japan
| |
Collapse
|
386
|
Malyshev IY, Budanova OP, Kuznetsova LV. Tumour adaptation to immune factors: old and new ideas for cancer immunotherapy. Can J Physiol Pharmacol 2023; 101:548-553. [PMID: 37728163 DOI: 10.1139/cjpp-2023-0083] [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: 09/21/2023]
Abstract
The tumour is fully functional in the zone of action of immune mediators. Moreover, the tumour needs immune system mediators to survive. "Adaptation" refers to a tumour's ability to withstand the effect of harmful elements. This gives birth to a new form of antitumour therapy: blocking tumour adaptability pathways. In this review, we will look at (i) tumour adaptation mechanisms as a result of pro-tumour immunoediting, (ii) how understanding tumour-adaptive mechanisms has led to ideas for developing cancer immunotherapies, and (iii) prospects for using the adaptation theory to substantiate new approaches to tumour growth inhibition. By considering the cancer problem through the lens of adaptability, a unique strategy for enhancing the efficacy of immunotherapy was proposed. The new approach is to utilise antisense treatment to erase the structural trace of adaptation in tumour cells or to disadapt tumour cells by "turning off" the immune system before initiating immunotherapy.
Collapse
Affiliation(s)
- I Yu Malyshev
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Moscow 127473, Russian Federation
- Federal State Budgetary Scientific Institution "Institute of general pathology and pathophysiology," Moscow 125315, Russian Federation
| | - O P Budanova
- Federal State Budgetary Scientific Institution "Institute of general pathology and pathophysiology," Moscow 125315, Russian Federation
| | - L V Kuznetsova
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Moscow 127473, Russian Federation
| |
Collapse
|
387
|
Li D, Wang D, Johann DJ, Hong H, Xu J. Assessments of tumor mutational burden estimation by targeted panel sequencing: A comprehensive simulation analysis. Exp Biol Med (Maywood) 2023; 248:1918-1926. [PMID: 38062992 PMCID: PMC10798187 DOI: 10.1177/15353702231211882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 01/06/2024] Open
Abstract
Tumor mutational burden (TMB), when at a high level, is an emerging indicative factor of sensitivity to immune checkpoint inhibitors. Previous studies have shown that the more affordable and accurate targeted panels can be used to measure TMB as a substitute for whole exome sequencing (WES). However, additional processes, such as hotspot mutations exclusion and TMB adjustment, are usually required to deal with the effect of the limited panel sizes. A comprehensive investigation of the effective factors is needed for accurate TMB estimation by targeted panels. In this study, we quantitatively evaluated the variances of TMB values calculated by WES and targeted panels using 10,000 simulated targeted panels with panel sizes ranging from 0.2 to 3.1 million bases. With The Cancer Genome Atlas (TCGA) cancer samples and mutation profiles, we fixed regressions on WES-TMBs and panel-TMBs to assess the performance of a given targeted panel. Panel size was found as one of the major effective factors of TMB estimation. Meanwhile, by investigating the well-performing small panels that reported TMB values similar to those of WES, we demonstrated the evidence of the cancer type-specific impacts of genes on TMB estimation and identified high-impact gene sets for different cancer types based on the TCGA data. This study revealed the quantitative correlations between TMB variance and panel size, and the potential impacts of individual genes on TMB estimation. Our results suggested that for cancer patients diagnosed using targeted panels, it would be highly beneficial to have the capability to directly measure TMB from the targeted sequencing data. This would greatly assist in making decisions regarding the use of immunotherapies.
Collapse
Affiliation(s)
- Dan Li
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR 72079, USA
| | - Dong Wang
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR 72079, USA
| | - Donald J Johann
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR 72079, USA
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
388
|
Ashitha KC, M G, N.R S, S B, P R. Leveraging mesoporous silica nanomaterial for optimal immunotherapeutics against cancer. IN VITRO MODELS 2023; 2:153-169. [PMID: 39872169 PMCID: PMC11756482 DOI: 10.1007/s44164-023-00061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/14/2023] [Accepted: 10/30/2023] [Indexed: 01/29/2025]
Abstract
Cancer represents a significant cause of morbidity and mortality. Definitive chemotherapy, surgery and radiotherapy treatment have not improved the "5-year survival period" and have shown recurrence. Currently, cancer immunotherapy is reported to be a promising therapeutic modality that aims to potentiate immune response against cancer by employing immune checkpoint inhibitors, cancer vaccines and immunomodulators. Inhibition of immune checkpoints such as PD-1/PDL1, CTLA and TIM molecules using monoclonal antibodies, ligands or both are proven to be the most successful anticancer immunotherapy. But the application of immunotherapy involves critical challenges such as non-responsiveness and systemic toxicity due to the administration of high dose. To mitigate the above challenges, nanomaterial-based delivery and therapy have been adopted to inhibit the immune checkpoints and induce an anticancer immune response. Specifically, mesoporous silica-based materials for cancer therapy are shown to be versatile materials for the above purpose. Mesoporous silica nanoparticle (MSN) based cancer immunotherapy overcomes numerous challenges and offers novel strategies for improving conventional immunotherapies. MSN has a high surface area, porosity and biocompatibility; it also has natural immune-adjuvant properties, which have been reported to be the best candidate material for immunotherapeutic delivery. This review will focus on the use of MSN as carriers for delivering immune checkpoint inhibitors and their efficacy in cancer combination therapy.
Collapse
Affiliation(s)
- K. C. Ashitha
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, 600025 India
| | - Gopinath M
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, 600025 India
| | - Sasirekha N.R
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, 600025 India
| | - Balakumar S
- National Centre for Nanoscience and Nanotechnology, University of Madras, Chennai, India
| | - Rajashree P
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, 600025 India
| |
Collapse
|
389
|
Huang KW, Huang TL. Association between programmed death-1 pathway and major depression. World J Biol Psychiatry 2023; 24:822-828. [PMID: 37139744 DOI: 10.1080/15622975.2023.2209876] [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: 02/16/2023] [Accepted: 04/30/2023] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Major depression (MD) may be associated with inflammation and immunity. PD-1 (programmed death-1), PD-L1 (programmed death-ligand 1) and PD-L2 (programmed death-ligand 2) are among the inhibitory immune mediators on the PD-1 pathway. However, previous data regarding the association between MD and PD-1 pathway were still scarce; therefore, we investigated the association of PD-1 pathway with MD. METHODS During a period of 2 years, patients with MD and healthy controls were recruited from a medical centre in this study. The diagnosis of MD was established according to the DSM-5 criteria. The severity of MD was assessed with 17-item Hamilton Depression Rating Scale. PD-1, PD-L1 and PD-L2 were detected in peripheral blood from MD patients after 4 weeks of treatment with antidepressant drugs. RESULTS A total of 54 patients with MD and 38 healthy controls were recruited. According to the analyses, there is a significantly higher PD-L2 level in MD than in healthy controls and lower PD-1 level after age and BMI adjustment. Besides, moderately positive correlation between HAM-D scores and PD-L2 level was found. CONCLUSIONS It was found that PD-1 pathway might play an important role in MD. We need a large sample to prove these results in the future.
Collapse
Affiliation(s)
- Kuan-Wei Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tiao-Lai Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Genomic and Proteomic Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
390
|
Lv M, Guo S, Zhang X, Zou Y, Chen Q, Zang C, Huang S, Hu Y, Wang Y, Wang Q, Zhong J. Attenuated Salmonella-delivered PD-1 siRNA enhances the antitumor effects of EZH2 inhibitors in colorectal cancer. Int Immunopharmacol 2023; 124:110918. [PMID: 37708707 DOI: 10.1016/j.intimp.2023.110918] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
Immunotherapy has made significant progress in the treatment of malignant tumors. However, strategies to combine immunotherapy with anticancer drugs have attracted great attention due to the low response rate and unique toxicity profile of immunotherapies and the subsequent development of acquired resistance in some initial responders. EZH2, a histone methyl transferase subunit of a Polycomb repressor complex,is highly expressed in a variety of tumors, and targeting EZH2 has become a new strategy for tumor therapy and drug combination. Here,we studied the effect of EZH2 inhibitors on colorectal cancer cells and their combination with immunotherapy. Our results demonstrated that EZH2 inhibitors can not only significantly inhibit the survival of colorectal cancer (CRC) cells and induce apoptosis, effectively inhibit cell invasion and migration, but also cause an increase in the expression of PD-L1 receptors on the cell surface. To determine the effect of EZH2 in combination with immunotherapy, we combine EZH2 inhibitors with PD-1 siRNA delivered by attenuated Salmonella. The vivo experiments have shown that the combination of EZH2 inhibitors and Salmonella-delivered PD-1 siRNA can further inhibit the development of CRC, trigger effective anti-tumor immunity, and improve therapeutic efficacy. Its underlying mechanisms mainly involve synergistic immunomodulation and apoptosis. This study suggests an emerging strategy based on a combination of EZH2 inhibitor and immunotherapy based on PD-1 inhibition.
Collapse
Affiliation(s)
- Mengmeng Lv
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Sheng Guo
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China, Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xinyu Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yan Zou
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Qiang Chen
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chongyi Zang
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Shuo Huang
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yuhan Hu
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yanling Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Qianqing Wang
- Department of Gynecology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, China.
| | - Jiateng Zhong
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China; Department of Gynecology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
391
|
Zhong W, Lu Y, Han X, Yang J, Qin Z, Zhang W, Yu Z, Wu B, Liu S, Xu W, Zheng C, Schuchter LM, Karakousis GC, Mitchell TC, Amaravadi R, Flowers AJ, Gimotty PA, Xiao M, Mills G, Herlyn M, Dong H, Mitchell MJ, Kim J, Xu X, Guo W. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep 2023; 42:113224. [PMID: 37805922 PMCID: PMC10697782 DOI: 10.1016/j.celrep.2023.113224] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 06/15/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages play a pivotal role in tumor immunity. We report that reprogramming of macrophages to tumor-associated macrophages (TAMs) promotes the secretion of exosomes. Mechanistically, increased exosome secretion is driven by MADD, which is phosphorylated by Akt upon TAM induction and activates Rab27a. TAM exosomes carry high levels of programmed death-ligand 1 (PD-L1) and potently suppress the proliferation and function of CD8+ T cells. Analysis of patient melanoma tissues indicates that TAM exosomes contribute significantly to CD8+ T cell suppression. Single-cell RNA sequencing analysis showed that exosome-related genes are highly expressed in macrophages in melanoma; TAM-specific RAB27A expression inversely correlates with CD8+ T cell infiltration. In a murine melanoma model, lipid nanoparticle delivery of small interfering RNAs (siRNAs) targeting macrophage RAB27A led to better T cell activation and sensitized tumors to anti-programmed cell death protein 1 (PD-1) treatment. Our study demonstrates tumors use TAM exosomes to combat CD8 T cells and suggests targeting TAM exosomes as a potential strategy to improve immunotherapies.
Collapse
Affiliation(s)
- Wenqun Zhong
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Youtao Lu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingbo Yang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiyuan Qin
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zhang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ziyan Yu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Wu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shujing Liu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathy Zheng
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn M Schuchter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahron J Flowers
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phyllis A Gimotty
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Gordon Mills
- Division of Oncological Science, School of Medicine, and Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Haidong Dong
- Departments of Urology and Immunology, Mayo College of Medicine and Science, Rochester, MN 55905, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhyong Kim
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
392
|
Ji S, Liu H, Pachella L, Stephenson RD, Groisberg R, Weiss SA. Use of immune checkpoint inhibitors in solid organ transplant recipients with advanced cutaneous malignancies. FRONTIERS IN TRANSPLANTATION 2023; 2:1284740. [PMID: 38993910 PMCID: PMC11235332 DOI: 10.3389/frtra.2023.1284740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/13/2023] [Indexed: 07/13/2024]
Abstract
Background Immune checkpoint inhibitors (ICI) are standard of care therapy for patients with cutaneous malignancies, the most frequently diagnosed cancers in solid organ transplant (SOT) recipients. The activity and rate of allograft rejection in SOT recipients with advanced skin cancers treated with ICI is understudied. Methods We conducted a retrospective analysis of SOT recipients with advanced melanoma, cutaneous squamous cell carcinoma (cSCC), and merkel cell carcinoma (MCC) who were treated with ICI. Unpublished cases from our institution and published cases from the literature were aggregated. Demographics, type of immunosuppressive therapy, type of ICI(s) administered, prior systemic therapies, tumor response to ICI, and evidence of organ rejection and/or failure were recorded. Objective response rates (ORR) and rates of graft rejection and failure are reported. Results Ninety patients were identified; four patients from our institution and 86 unique patients from a literature review. ORR to first-line ICI for the entire cohort was 41.1% (37/90). ORR by tumor type was 31% (18/58), 64.3% (18/28), and 25.0% (1/4) for melanoma, cSCC, and MCC, respectively. The rate of graft rejection was 37.8% (34/90) with 61.8% (21/34) of these cases progressing to graft failure. Number of immunosuppressive agents (0, 1, 2, or 3) was inversely associated with rate of graft failure. Conclusions In this retrospective analysis, ICIs demonstrate clinical activity in SOT recipients with cutaneous malignancies; however, the rate of graft rejection is high. Treatment plans should be individualized through thorough interdisciplinary discussion. Immunosuppressive modifications may be considered prior to starting treatment, but when feasible, enrollment on clinical trials is preferred.
Collapse
Affiliation(s)
- Stephanie Ji
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, New Brunswick, NJ, United States
| | - Laura Pachella
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Ryan D Stephenson
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Roman Groisberg
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Sarah A Weiss
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
393
|
Diab A, Gogas H, Sandhu S, Long GV, Ascierto PA, Larkin J, Sznol M, Franke F, Ciuleanu TE, Pereira C, Muñoz Couselo E, Bronzon Damian F, Schenker M, Perfetti A, Lebbe C, Quéreux G, Meier F, Curti BD, Rojas C, Arriaga Y, Yang H, Zhou M, Ravimohan S, Statkevich P, Tagliaferri MA, Khushalani NI. Bempegaldesleukin Plus Nivolumab in Untreated Advanced Melanoma: The Open-Label, Phase III PIVOT IO 001 Trial Results. J Clin Oncol 2023; 41:4756-4767. [PMID: 37651676 PMCID: PMC10602507 DOI: 10.1200/jco.23.00172] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/23/2023] [Accepted: 06/29/2023] [Indexed: 09/02/2023] Open
Abstract
PURPOSE Despite marked advances in the treatment of unresectable or metastatic melanoma, the need for novel therapies remains. Bempegaldesleukin (BEMPEG), a pegylated interleukin-2 (IL-2) cytokine prodrug, demonstrated efficacy in the phase II PIVOT-02 trial. PIVOT IO 001 (ClinicalTrials.gov identifier: NCT03635983) is a phase III, randomized, open-label study that builds on the PIVOT-02 results in first-line melanoma. METHODS Patients with previously untreated, unresectable, or metastatic melanoma were randomly assigned 1:1 to receive BEMPEG plus nivolumab (NIVO) or NIVO monotherapy. Primary end points were objective response rate (ORR) and progression-free survival (PFS) by blinded independent central review and overall survival (OS). Secondary and exploratory end points included additional efficacy measures, safety, and pharmacokinetics (PKs) and pharmacodynamics analyses. RESULTS In 783 patients (n = 391, BEMPEG plus NIVO; n = 392, NIVO monotherapy), the median follow-up was 11.6 months in the intent-to-treat population. The ORR with BEMPEG plus NIVO was 27.7% versus 36.0% with NIVO (two-sided P = .0311). The median PFS with BEMPEG plus NIVO was 4.17 months (95% CI, 3.52 to 5.55) versus 4.99 months (95% CI, 4.14 to 7.82) with NIVO (hazard ratio [HR], 1.09; 97% CI, 0.88 to 1.35; P = .3988). The median OS was 29.67 months (95% CI, 22.14 to not reached [NR]) with BEMPEG plus NIVO versus 28.88 months (95% CI, 21.32 to NR) with NIVO (HR, 0.94; 99.929% CI, 0.59 to 1.48; P = .6361). Grade 3-4 treatment-related adverse events (AEs) and serious AE rates were higher with the combination (21.7% and 10.1%, respectively) versus NIVO (11.5% and 5.5%, respectively). BEMPEG PK exposure and absolute lymphocyte count changes after BEMPEG plus NIVO were comparable between PIVOT IO 001 and PIVOT-02. CONCLUSION The PIVOT IO 001 study did not meet its primary end points of ORR, PFS, and OS. Increased toxicity was observed with BEMPEG plus NIVO versus NIVO.
Collapse
Affiliation(s)
- Adi Diab
- Melanoma Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Shahneen Sandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
| | - Georgina V. Long
- Melanoma Institute Australia, Royal North Shore and Mater Hospitals, The University of Sydney, Sydney, NSW, Australia
| | - Paolo A. Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Department, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - James Larkin
- Medical Oncology, The Royal Marsden Hospital, London, United Kingdom
| | - Mario Sznol
- Medical Oncology, Yale Cancer Center, Yale University School of Medicine, Smilow Cancer Hospital Yale New Haven Health, New Haven, CT
| | - Fabio Franke
- Medical Oncology, Oncosite Centro de Pesquisa Clínica, Ijui, Brazil
| | - Tudor E. Ciuleanu
- Medical Oncology, Institutul Prof Dr Ion Chiricuţă, Cluj-Napoca, Romania
| | - Caio Pereira
- Fundação Pio XII, Hospital de Câncer de Barretos, Barretos, Brazil
| | - Eva Muñoz Couselo
- Medical Oncology, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Michael Schenker
- Sf Nectarie Oncology Center, University of Medicine and Pharmacy, Craiova, Romania
| | - Aldo Perfetti
- Clínica Adventista Belgrano, Buenos Aires, Argentina
| | - Celeste Lebbe
- AP-HP Department of Dermato-oncology and CIC, INSERM U976, Cancer Institute APHP, Nord-Université Paris Cite, Université Paris Cité, Paris, France
| | - Gaëlle Quéreux
- Department of Dermatology, CIC 1413, de Cancéro-Dermatologie-CIC Biothérapie Nantes, Nantes University Hospital, Nantes, France
| | - Friedegund Meier
- Skin Cancer Center, National Center for Tumor Diseases, University Cancer Centre Dresden, Dresden, Germany
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Brendan D. Curti
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR
| | - Carlos Rojas
- Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
| | - Yull Arriaga
- Medical Oncology, Bristol Myers Squibb, Princeton, NJ
| | - Haisu Yang
- Medical Oncology, Bristol Myers Squibb, Princeton, NJ
| | - Ming Zhou
- Medical Oncology, Bristol Myers Squibb, Princeton, NJ
| | | | - Paul Statkevich
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ
| | | | | |
Collapse
|
394
|
Zheng H, Wang G, Wang Y, Liu J, Ma G, Du J. Systematic analysis reveals a pan-cancer SNHG family signature predicting prognosis and immunotherapy response. iScience 2023; 26:108055. [PMID: 37854704 PMCID: PMC10579433 DOI: 10.1016/j.isci.2023.108055] [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: 04/24/2023] [Revised: 08/16/2023] [Accepted: 09/22/2023] [Indexed: 10/20/2023] Open
Abstract
Small nucleolar RNA host genes (SNHGs) are a special family of long non-coding RNAs (lncRNAs), which not only function in a way similar to other lncRNAs but also influence the intracellular level of small nucleolar RNAs to modulate cancers. However, the features of SNHGs and their role in the prognosis and immunotherapeutic response of human cancer have not been explored. We found that SNHGs were commonly deregulated and correlated with patient survival in various cancers. The critical role of DNA methylation and somatic alterations on deregulation was also identified. SNHG family score was significantly associated with survival, multiple tumor characteristics, and tumor microenvironment. SNHG-related risk score could serve as a prognostic and immunotherapeutic response biomarker based on multiple databases. This study emphasizes the potential of SNHGs as biomarkers for prognosis and immunotherapeutic response, enabling further research into the immune regulatory mechanism and therapeutic potentials of SNHGs in cancer.
Collapse
Affiliation(s)
- Haotian Zheng
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Guanghui Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Yadong Wang
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Jichang Liu
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Guoyuan Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Thoracic Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
395
|
Peisen F, Gerken A, Hering A, Dahm I, Nikolaou K, Gatidis S, Eigentler TK, Amaral T, Moltz JH, Othman AE. Can Whole-Body Baseline CT Radiomics Add Information to the Prediction of Best Response, Progression-Free Survival, and Overall Survival of Stage IV Melanoma Patients Receiving First-Line Targeted Therapy: A Retrospective Register Study. Diagnostics (Basel) 2023; 13:3210. [PMID: 37892030 PMCID: PMC10605712 DOI: 10.3390/diagnostics13203210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate whether the combination of radiomics and clinical parameters in a machine-learning model offers additive information compared with the use of only clinical parameters in predicting the best response, progression-free survival after six months, as well as overall survival after six and twelve months in patients with stage IV malignant melanoma undergoing first-line targeted therapy. METHODS A baseline machine-learning model using clinical variables (demographic parameters and tumor markers) was compared with an extended model using clinical variables and radiomic features of the whole tumor burden, utilizing repeated five-fold cross-validation. Baseline CTs of 91 stage IV malignant melanoma patients, all treated in the same university hospital, were identified in the Central Malignant Melanoma Registry and all metastases were volumetrically segmented (n = 4727). RESULTS Compared with the baseline model, the extended radiomics model did not add significantly more information to the best-response prediction (AUC [95% CI] 0.548 (0.188, 0.808) vs. 0.487 (0.139, 0.743)), the prediction of PFS after six months (AUC [95% CI] 0.699 (0.436, 0.958) vs. 0.604 (0.373, 0.867)), or the overall survival prediction after six and twelve months (AUC [95% CI] 0.685 (0.188, 0.967) vs. 0.766 (0.433, 1.000) and AUC [95% CI] 0.554 (0.163, 0.781) vs. 0.616 (0.271, 1.000), respectively). CONCLUSIONS The results showed no additional value of baseline whole-body CT radiomics for best-response prediction, progression-free survival prediction for six months, or six-month and twelve-month overall survival prediction for stage IV melanoma patients receiving first-line targeted therapy. These results need to be validated in a larger cohort.
Collapse
Affiliation(s)
- Felix Peisen
- Department of Diagnostic and Interventional Radiology, Tuebingen University Hospital, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.); (A.E.O.)
| | - Annika Gerken
- Fraunhofer MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany; (A.G.); (A.H.); (J.H.M.)
| | - Alessa Hering
- Fraunhofer MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany; (A.G.); (A.H.); (J.H.M.)
- Diagnostic Image Analysis Group, Radboud University Medical Center (Radboudumc), Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Isabel Dahm
- Department of Diagnostic and Interventional Radiology, Tuebingen University Hospital, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.); (A.E.O.)
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Tuebingen University Hospital, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.); (A.E.O.)
- Image-Guided and Functionally Instructed Tumor Therapies (iFIT), The Cluster of Excellence (EXC 2180), 72076 Tuebingen, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Tuebingen University Hospital, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.); (A.E.O.)
- Max Planck Institute for Intelligent Systems, Max-Planck-Ring 4, 72076 Tuebingen, Germany
| | - Thomas K. Eigentler
- Center of Dermato-Oncology, Department of Dermatology, Tuebingen University Hospital, Eberhard Karls University, Liebermeisterstraße 25, 72076 Tuebingen, Germany; (T.K.E.); (T.A.)
- Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humbolt-Universität zu Berlin, Luisenstraße 2, 10117 Berlin, Germany
| | - Teresa Amaral
- Center of Dermato-Oncology, Department of Dermatology, Tuebingen University Hospital, Eberhard Karls University, Liebermeisterstraße 25, 72076 Tuebingen, Germany; (T.K.E.); (T.A.)
| | - Jan H. Moltz
- Fraunhofer MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany; (A.G.); (A.H.); (J.H.M.)
| | - Ahmed E. Othman
- Department of Diagnostic and Interventional Radiology, Tuebingen University Hospital, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany; (I.D.); (K.N.); (S.G.); (A.E.O.)
- Institute of Neuroradiology, Johannes Gutenberg University Hospital Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
396
|
Modesto A, Tougeron D, Tremolières P, Ronchin P, Jouve AD, Leignel DA, Vendrely V, Riou O, Martin-Babau J, Le Sourd S, Mirabel X, Leroy T, Huguet F, Montaigne L, Baumgaertner I, Deslandres M, Moyal E, Seva C, Selves J, Otal P, Pezzella V, Guimbaud R, Filleron T, Quéro L. Association of Radiochemotherapy to Immunotherapy in unresectable locally advanced Oesophageal carciNoma-randomized phase 2 trial ARION UCGI 33/PRODIGE 67: the study protocol. BMC Cancer 2023; 23:966. [PMID: 37828434 PMCID: PMC10568784 DOI: 10.1186/s12885-023-11227-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/25/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND In case of locally advanced and/or non-metastatic unresectable esophageal cancer, definitive chemoradiotherapy (CRT) delivering 50 Gy in 25 daily fractions in combination with platinum-based regimen remains the standard of care resulting in a 2-year disease-free survival of 25% which deserves to be associated with new systemic strategies. In recent years, several immune checkpoint inhibitors (anti-PD1/anti-PD-L1, anti-Program-Death 1/anti-Program-Death ligand 1) have been approved for the treatment of various solid malignancies including metastatic esophageal cancer. As such, we hypothesized that the addition of an anti-PD-L1 to CRT would provide clinical benefit for patients with locally advanced oesophageal cancer. To assess the efficacy of the anti-PD-L1 durvalumab in combination with CRT and then as maintenance therapy we designed the randomized phase II ARION (Association of Radiochemotherapy with Immunotherapy in unresectable Oesophageal carciNoma- UCGI 33/PRODIGE 67). METHODS ARION is a multicenter, open-label, randomized, comparative phase II trial. Patients are randomly assigned in a 1:1 ratio in each arm with a stratification according to tumor stage, histology and centre. Experimental arm relies on CRT with 50 Gy in 25 daily fractions in combination with FOLFOX regimen administrated during and after radiotherapy every two weeks for a total of 6 cycles and durvalumab starting with CRT for a total of 12 infusions. Standard arm is CRT alone. Use of Intensity Modulated radiotherapy is mandatory. The primary endpoint is to increase progression-free survival at 12 months from 50 to 68% (HR = 0.55) (power 90%; one-sided alpha-risk, 10%). Progression will be defined with central external review of imaging. ANCILLARY STUDIES ARE PLANNED PD-L1 Combined Positivity Score on carcinoma cells and stromal immune cells of diagnostic biopsy specimen will be correlated to disease free survival. The study of gut microbiota will aim to determine if baseline intestinal bacteria correlates with tumor response. Proteomic analysis on blood samples will compare long-term responder after CRT with durvalumab to non-responder to identify biomarkers. CONCLUSION Results of the present study will be of great importance to evaluate the impact of immunotherapy in combination with CRT and decipher immune response in this unmet need clinical situation. TRIAL REGISTRATION ClinicalTrials.gov, NCT: 03777813.Trial registration date: 5th December 2018.
Collapse
Affiliation(s)
- Anouchka Modesto
- Radiation Oncology Department Institut Claudius Regaud at Institut, Universitaire du Cancer de Toulouse-Oncopole, 1 Rue Irene Joliot Curie, 31059, Toulouse, France.
- Inserm Team 11 RadOpt CRCT 1, Avenue Hubert Curien, 31059, Toulouse, France.
| | - David Tougeron
- Service d'hépato-Gastro-Entérologie, Centre Hospitalier Universitaire de Poitiers, 86000, Poitiers, France
| | - Pierre Tremolières
- Institut de Cancérologie de L'Ouest: Angers Et Saint Herblain, Saint-herblain, France
| | - Philippe Ronchin
- Hôpital Privé Arnault Tzanck- Centre Azuréen de Cancérologie, Mougins, France
| | | | | | | | | | | | | | | | - Thomas Leroy
- Nouvelle Clinique Des Dentellières, Valenciennes, France
| | - Florence Huguet
- Radiation Oncology Department, Tenon Hospital, AP-HP,, Sorbonne University, Paris, France
| | | | | | - Marion Deslandres
- GI Oncology Department Centre Hospitalo, Universitaire Rangueil, Toulouse, France
| | - Elizabeth Moyal
- Radiation Oncology Department Institut Claudius Regaud at Institut, Universitaire du Cancer de Toulouse-Oncopole, 1 Rue Irene Joliot Curie, 31059, Toulouse, France
- Inserm Team 11 RadOpt CRCT 1, Avenue Hubert Curien, 31059, Toulouse, France
| | - Catherine Seva
- Inserm Team 11 RadOpt CRCT 1, Avenue Hubert Curien, 31059, Toulouse, France
| | - Janick Selves
- Pathology department, Centre Hospitalo Universitaire IUCT-Oncopole, Toulouse, France
| | - Philippe Otal
- Imaging Department Centre Hospitalo, Universitaire Rangueil, Toulouse, France
| | | | - Rosine Guimbaud
- GI Oncology Department Centre Hospitalo, Universitaire Rangueil, Toulouse, France
| | - Thomas Filleron
- Biostatistics Departement Institut Claudius Regaud Institut, Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Laurent Quéro
- Radiation Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| |
Collapse
|
397
|
Huang J, Gong C, Zhou A. Modulation of gut microbiota: a novel approach to enhancing the effects of immune checkpoint inhibitors. Ther Adv Med Oncol 2023; 15:17588359231204854. [PMID: 37841750 PMCID: PMC10571694 DOI: 10.1177/17588359231204854] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have greatly improved the prognosis of some cancer patients, the majority still fail to respond adequately, and the available biomarkers cannot reliably predict drug efficacy. The gut microbiota has received widespread attention among the various intrinsic and extrinsic factors contributing to drug resistance. As an essential regulator of physiological function, the impact of gut microbiota on host immunity and response to cancer therapy is increasingly recognized. Several studies have demonstrated significant differences in gut microbiota between responders and nonresponders. The gut microbiota associated with better clinical outcomes is called 'favorable gut microbiota'. Significantly, interventions can alter the gut microbiota. By shifting the gut microbiota to the 'favorable' one through various modifications, preclinical and clinical studies have yielded more pronounced responses and better clinical outcomes when combined with ICIs treatment, providing novel approaches to improve the efficacy of cancer immunotherapy. These findings may be attributed to the effects of gut microbiota and its metabolites on the immune microenvironment and the systemic immune system, but the underlying mechanisms remain to be discovered. In this review, we summarize the clinical evidence that the gut microbiota is strongly associated with the outcomes of ICI treatment and describe the gut microbiota characteristics associated with better clinical outcomes. We then expand on the current prevalent modalities of gut microbiota regulation, provide a comprehensive overview of preclinical and clinical research advances in improving the therapeutic efficacy and prognosis of ICIs by modulating gut microbiota, and suggest fundamental questions we need to address and potential directions for future research expansion.
Collapse
Affiliation(s)
- Jinglong Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100020, China
| |
Collapse
|
398
|
Liang J, Vitale T, Zhang X, Jackson TD, Yu D, Jedrychowski M, Gygi SP, Widlund HR, Wucherpfennig KW, Puigserver P. Selective Mitochondrial Respiratory Complex I Subunit Deficiency Causes Tumor Immunogenicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560316. [PMID: 37873273 PMCID: PMC10592908 DOI: 10.1101/2023.10.02.560316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Targeting of specific metabolic pathways in tumor cells has the potential to sensitize them to immune-mediated attack. Here we provide evidence for a specific means of mitochondrial respiratory Complex I (CI) inhibition that improves tumor immunogenicity and sensitivity to immune checkpoint blockade (ICB). Targeted genetic deletion of the CI subunits Ndufs4 and Ndufs6 , but not other subunits, induces an immune-dependent tumor growth attenuation in mouse melanoma models. We show that deletion of Ndufs4 induces expression of the transcription factor Nlrc5 and genes in the MHC class I antigen presentation and processing pathway. This induction of MHC-related genes is driven by an accumulation of pyruvate dehydrogenase-dependent mitochondrial acetyl-CoA downstream of CI subunit deletion. This work provides a novel functional modality by which selective CI inhibition restricts tumor growth, suggesting that specific targeting of Ndufs4 , or related CI subunits, increases T-cell mediated immunity and sensitivity to ICB.
Collapse
|
399
|
Chamoto K, Yaguchi T, Tajima M, Honjo T. Insights from a 30-year journey: function, regulation and therapeutic modulation of PD1. Nat Rev Immunol 2023; 23:682-695. [PMID: 37185300 DOI: 10.1038/s41577-023-00867-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 05/17/2023]
Abstract
PD1 was originally discovered in 1992 as a molecule associated with activation-induced cell death in T cells. Over the past 30 years, it was found that PD1 has a critical role in avoiding overactivation-induced cell death and autoimmunity, whereas its inhibition unleashes anticancer immunity. Here, we outline the journey from the discovery of PD1 to its role as a breakthrough target in cancer immunotherapy. We describe its regulation and function and examine how a mechanistic understanding of PD1 signalling suggests a central function in setting the T cell activation threshold, thereby controlling T cell proliferation, differentiation, exhaustion and metabolic status. This threshold theory, in combination with new insights into T cell metabolism and a better understanding of immune cell modulation by the microbiota, can provide guidance for the development of efficient combination therapies. Moreover, we discuss the mechanisms underlying immune-related adverse events after PD1-targeted therapy and their possible treatment.
Collapse
Affiliation(s)
- Kenji Chamoto
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomonori Yaguchi
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Tajima
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tasuku Honjo
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
400
|
Soerensen AV, Kjellberg J, Ibsen R, Bastholt L, Schmidt H, Svane IM. Health care and socioeconomic costs for long-term survivors after implementation of checkpoint-inhibitors and targeted agents for metastatic melanoma. Eur J Cancer 2023; 192:113288. [PMID: 37672816 DOI: 10.1016/j.ejca.2023.113288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Real-life data on health care costs and loss of productivity after implementing new agents for metastatic melanoma are important to supplement model-based economic data. MATERIALS AND METHODS All patients registered in the Danish Metastatic Melanoma Database (DAMMED) and the National Patient Registry in 2007-2011 were compared to 2012-2016 after the implementation of checkpoint inhibitors and targeted therapy. Health care costs, social transfer income (STI), and loss of productivity were calculated with a 2-step one model generalised linear regression (GLM) model. Medicine costs were calculated separately. RESULTS In 2007-2011, 70 (15%) out of 464 patients were long-term survivors compared to 347 (32%) out of 1089 patients in 2012-2016. Total health care costs per patient year were significantly lower in the first treatment year (€41.457 versus €60.547, relative change (RC) 0.72, 95% confidence interval (CI) 0.56-0.94, p = 0.015) and without significant difference the second year in 2012-2016 compared to 2007-2011. Medicine costs per patient year increased the first (€85.464 versus €26.339, RC 3.39, 95% CI 2.61-4.41, p < 0.001) and the second (€26.464 versus €11.150, RC 2.59, 95% CI 1.98-3.40, p < 0.001) year in 2012-2016 compared to 2007-2011. Productivity increased for long-term survivors in 2012-2016 in contrast to 2007-2011. CONCLUSION Implementation of targeted therapy and checkpoint-inhibitors has increased medicine costs more than three-fold for long-term survivors. Total health care costs excluding medicine costs were significantly lower for long-term survivors the first and without change the second treatment year in 2012-2016 compared to 2007-2011. However, the number of treated patients increased which leads to an increase in overall total health care costs. Importantly, productivity increased for long-term survivors in 2012-2016.
Collapse
Affiliation(s)
- Anne Vest Soerensen
- Department of Oncology, Copenhagen University Hospital Herlev, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark.
| | - Jakob Kjellberg
- The Danish Center for Social Science Research (VIVE), Copenhagen, Herluf Trolles Gade 11, 1052 Copenhagen K, Denmark
| | - Rikke Ibsen
- i2Minds, Klosterport 4E 4., 8000 Aarhus, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, J.B. Winsloews vej 4, 5000 Odense C, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Borgmester Ib Juuls Vej 25C, 2730 Herlev, Denmark
| |
Collapse
|