1
|
Chen P, Zhang Y, Bordeau BM, Balthasar JP. Assessment of the Effects of Single-Domain Anti-Idiotypic Distribution Enhancers on the Disposition of Trastuzumab and on the Efficacy of a PE24-Trastuzumab Immunotoxin. Cancers (Basel) 2025; 17:1468. [PMID: 40361392 PMCID: PMC12071152 DOI: 10.3390/cancers17091468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Antibody-based therapies often exhibit limited distribution within solid tumors due to the "binding-site barrier" (BSB). Our group has developed and validated the use of anti-idiotypic distribution enhancers (AIDEs), which transiently block antibody binding, improving intra-tumoral distribution and efficacy. This study evaluated 1HE and LG1, model anti-trastuzumab AIDEs, in combination with trastuzumab-PE24, a highly potent immunotoxin. METHODS The effects of 1HE on the whole-body disposition of radiolabeled trastuzumab were assessed in NCI-N87 tumor-bearing mice. Mechanistic pharmacokinetic/pharmacodynamic (PK/PD) modeling was employed to explore how AIDE binding kinetics influence antibody intra-tumoral distribution and immunotoxin potency. Trastuzumab-PE24 was developed by site-specific conjugation, enabled by self-splicing split intein, with cytotoxicity tested on various cell lines in vitro. The impact of 1HE and LG1 coadministration on trastuzumab-PE24 efficacy was evaluated in NCI-N87 xenograft-bearing mice. RESULTS 1HE coadministration decreased trastuzumab tumor maximum concentration, reducing tumor terminal slope by 8% and overall tumor exposure by 2.6%, without negatively affecting selectivity. Modeling predicted the optimal AIDE dissociation rate constant for trastuzumab-PE24 to be between 0.015 and 0.3 h-1. The coadministration of trastuzumab-PE24 with 1HE and LG1 improved anti-tumor efficacy and extended median survival to 60 days (p = 0.0002). CONCLUSIONS AIDE coadministration led to minimal negative impacts on overall tumor exposure, consistent with model simulations. AIDE coadministration improved the efficacy of trastuzumab-PE24 in NCI-N87 xenografts. Modeling further predicted that repeated AIDE administration with trastuzumab-PE24 could induce complete tumor regression. These findings highlight the advantages of the AIDE strategy, particularly when coadministered with highly potent immunotoxins.
Collapse
Affiliation(s)
| | | | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA; (P.C.); (Y.Z.); (B.M.B.)
| |
Collapse
|
2
|
Kumari S. Mesothelin as a Signal Pathways and Epigenetic Target in Cancer Therapy. Cancers (Basel) 2025; 17:1118. [PMID: 40227616 PMCID: PMC11987799 DOI: 10.3390/cancers17071118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Mesothelin (MSLN), a glycoprotein-based tumor antigen, is elevated in several malignancies and it is related to a poor prognosis, as it enhances tumor aggression, dissemination and chemotherapy resistance. MSLN plays a crucial role in epigenetic and signal pathway regulation and it can be an important biomarker. MSLN targeting is in particular, associated with CA125/MUC16, which offers the potential to improve lung, pancreatic, colon and ovarian cancer detection as well as therapeutic strategies. MSLNtargeted therapies have shown favorable results, such as CAR NK cells, 227Th conjugate and CAR-T cells, which target mesothelin. Significant advancements can be achieved with novel techniques, such as mesothelin-targeting BiTEs and simultaneous CAR-T cells. Immunotherapies targeting mesothelin have the potential to completely transform the way cancer is therapy in patients with limited options. To fully comprehend the mechanisms of MSLN, more investigation is required to explore its role in cancer for improved patient outcomes. The complex control, cellular functions and clinical significance of MSLN in the advancement of cancer are highlighted in this review.
Collapse
Affiliation(s)
- Seema Kumari
- Department of Biotechnology, Dr. B.R Ambedkar University, Srikakulam 532410, Andhra Pradesh, India
| |
Collapse
|
3
|
Mir S, Venugopalan A, Zhang J, Nair NU, Sengupta M, Khanal M, Stathopoulou C, Jiang Q, Hassan R. Persistence of activated anti-mesothelin hYP218 chimeric antigen receptor T cells in the tumour is associated with efficacy in gastric and colorectal carcinomas. Clin Transl Med 2024; 14:e70057. [PMID: 39548594 PMCID: PMC11567854 DOI: 10.1002/ctm2.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 11/18/2024] Open
Abstract
Patients with advanced gastric and colorectal cancers have limited treatment options. Since mesothelin is highly expressed in these tumour types, we evaluated the therapeutic benefits of anti-mesothelin hYP218 CAR T cells alone, and in combination with anti-PD1 antibody, pembrolizumab. GEPIA analysis was performed using human gastric (n = 408) and colon cancer tumours (n = 275) in TCGA database, to evaluate mRNA expression of mesothelin, compared to normal tissues. Mesothelin expression in gastric and colorectal cancer cell-lines (n = 5) was analysed using flow cytometry. In vitro efficacy by hYP218 CAR T cells was tested by cytotoxicity and cytokine release assays. In vivo anti-tumour efficacy of hYP218 CAR T cells alone, and in combination with pembrolizumab, was evaluated in NSG mice bearing human gastric (HGC27) and colorectal (SW48) tumour xenografts. Additionally, hYP218 CAR-T cell persistence, activation and exhaustion marker-expression were studied. Mesothelin expression was significantly higher in gastric and colon cancer biopsies compared to normal tissues (p < .005). Mesothelin expression in gastric and colon cancer cell lines ranged from 10 000 to 70 000 molecules per cell. hYP218 CAR T cells demonstrated strong cytotoxic activity at low effector to target ratio, ranging from 0.24 to 1.0. In NSG mouse-models, hYP218 CAR T cells demonstrated anti-tumour efficacy and persisted in the tumour microenvironment in a functional state at day 40 posttreatment with expression of activation markers CD39 and CD69, increased production of IFN-γ and TNF-α and ability to kill tumour cells in vitro when isolated from tumours. There was increased PD1 expression. In combination with pembrolizumab, hYP218 CAR T cells led to slower tumour growth in NSG mice bearing large but not small HGC27 tumours. Anti-tumour efficacy of hYP218 CAR T cells is due to increased accumulation of activated CAR T cells in the tumour and combination with pembrolizumab resulted in improvement in anti-tumour activity of large established tumours. HIGHLIGHTS: Mesothelin expression is significantly higher in gastric and colorectal cancers than normal tissues. hYP218 CAR T cells demonstrate strong anti-tumour activity against mesothelin-positive gastric and colorectal carcinomas. Activated hYP218 CAR T cells persist in the tumour microenvironment and retain their cytotoxic activity. Addition of pembrolizumab in larger tumours enhance CAR T cell efficacy.
Collapse
Affiliation(s)
- Sameer Mir
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Abhilash Venugopalan
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Jingli Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Nishanth Ulhas Nair
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Manjistha Sengupta
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Manakamana Khanal
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Chaido Stathopoulou
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Qun Jiang
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)BethesdaMarylandUSA
| |
Collapse
|
4
|
Mathiot L, Baldini C, Letissier O, Hollebecque A, Bahleda R, Gazzah A, Smolenschi C, Sakkal M, Danlos FX, Henon C, Beshiri K, Goldschmidt V, Parisi C, Patrikidou A, Michot JM, Marabelle A, Postel-Vinay S, Bernard-Tessier A, Loriot Y, Ponce S, Champiat S, Ouali K. Exploring the Role of Target Expression in Treatment Efficacy of Antibody-Drug Conjugates (ADCs) in Solid Cancers: A Comprehensive Review. Curr Oncol Rep 2024; 26:1236-1248. [PMID: 39066847 DOI: 10.1007/s11912-024-01576-9] [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] [Accepted: 06/29/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) offer a promising path for cancer therapy, leveraging the specificity of monoclonal antibodies and the cytotoxicity of linked drugs. The success of ADCs hinges on precise targeting of cancer cells based on protein expression levels. This review explores the relationship between target protein expression and ADC efficacy in solid tumours, focusing on results of clinical trials conducted between January 2019 and May 2023. RECENT FINDINGS We hereby highlight approved ADCs, revealing their effectiveness even in low-expressing target populations. Assessing target expression poses challenges, owing to variations in scoring systems and biopsy types. Emerging methods, like digital image analysis, aim to standardize assessment. The complexity of ADC pharmacokinetics, tumour dynamics, and off-target effects emphasises the need for a balanced approach. This review underscores the importance of understanding target protein dynamics and promoting standardized evaluation methods in shaping the future of ADC-based cancer therapies.
Collapse
Affiliation(s)
- Laurent Mathiot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Capucine Baldini
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Octave Letissier
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Antoine Hollebecque
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Rastislav Bahleda
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Anas Gazzah
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Cristina Smolenschi
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Madona Sakkal
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - François-Xavier Danlos
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Villejuif, France
- Centre d'Investigations Cliniques Biothérapies Pour Une Immunisation in Situ (BIOTHERIS), INSERM, CIC1428, Villejuif, France
| | - Clémence Henon
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Kristi Beshiri
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Vincent Goldschmidt
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Claudia Parisi
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Anna Patrikidou
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Jean-Marie Michot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Aurélien Marabelle
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Sophie Postel-Vinay
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | | | - Yohann Loriot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U981, Villejuif, France
| | - Santiago Ponce
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Stéphane Champiat
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Villejuif, France
- Centre d'Investigations Cliniques Biothérapies Pour Une Immunisation in Situ (BIOTHERIS), INSERM, CIC1428, Villejuif, France
| | - Kaïssa Ouali
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France.
| |
Collapse
|
5
|
Nabeta R, Kanaya A, Shimada K, Matsuura K, Yoshimura A, Oyamada T, Azakami D, Furuya T, Uchide T. Characterization of mesothelin gene expression in dogs and overexpression in canine mesotheliomas. Front Vet Sci 2024; 11:1436621. [PMID: 39315086 PMCID: PMC11417096 DOI: 10.3389/fvets.2024.1436621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Canine mesotheliomas are uncommon malignant tumors typically detected late. Minimally invasive diagnostic biomarkers would facilitate diagnosis at earlier stages, thereby improving clinical outcomes. We hypothesized that mesothelin could be used as a reliable diagnostic biomarker for canine mesotheliomas since it has been used as a cancer biomarker for human mesothelioma. We aimed to explore and characterize mesothelin gene expression in dogs and assess its use as a diagnostic biomarker for canine mesotheliomas. Materials and methods We quantified expressed canine mesothelin transcripts via reverse transcription polymerase chain reaction (RT-PCR) and sequenced them using ribonucleic acid (RNA) extracted from a canine mesothelioma cell line. After confirming mesothelin expression, we assessed its levels in major organ tissues and compared them with those in the mesothelioma tissues using quantitative PCR (qPCR). Mesothelin overexpression in mesotheliomas was detected, and we further compared its levels using qPCR between mesotheliomas and non-mesotheliomas using tumor tissues and clinical sample effusions, confirming its significance as a diagnostic biomarker for canine mesothelioma. Results Mesothelin complementary deoxyribonucleic acid (cDNA) was amplified via RT-PCR, yielding a single band of expected upon DNA electrophoresis. Sequence analyses confirmed it as a predicted canine mesothelin transcript from the genome sequence database. Comparative sequence analysis of the deduced amino acid sequence of the expressed canine mesothelin demonstrated molecular signature similarities with the human mesothelin. However, the pre-sequence of canine mesothelin lacks the mature megakaryocyte potentiating factor (MPF) portion, which is typically cleaved post-translationally with furin. Mesothelin expression was quantified via qPCR revealing low levels in the mesothelial and lung tissues, with negligible expression in the other major organs. Canine mesothelin exhibited significantly higher expression in the canine mesotheliomas than in the noncancerous tissues. Moreover, analysis of clinical samples using qPCR demonstrated markedly elevated mesothelin expression in canine mesotheliomas compared to non-mesothelioma cases. Discussion and conclusion Canine mesothelin exhibits molecular and biological characteristics akin to human mesothelin. It could serve as a vital biomarker for diagnosing canine mesotheliomas, applicable to both tissue- and effusion-based samples.
Collapse
Affiliation(s)
- Rina Nabeta
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Ami Kanaya
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Kazumi Shimada
- Laboratory of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Katsuhiro Matsuura
- Laboratory of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Small Animal Clinical Science, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Aritada Yoshimura
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tomohiro Oyamada
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Daigo Azakami
- Laboratory of Veterinary Clinical Oncoogy, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tetsuya Furuya
- Laboratory of Veterinary Infectious Diseases, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tsuyoshi Uchide
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
6
|
Huang M, Park J, Seo J, Ko S, Yang YH, Lee Y, Kim HJ, Lee BS, Lee YS, Ko BJ, Jung ST, Park D, Yoo TH, Kim CH. An epidermal growth factor receptor-targeting immunotoxin based on IgG shows potent antitumor activity against head and neck cancer. FASEB J 2024; 38:e23759. [PMID: 38949635 DOI: 10.1096/fj.202301968r] [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: 09/26/2023] [Revised: 05/18/2024] [Accepted: 06/13/2024] [Indexed: 07/02/2024]
Abstract
The epidermal growth factor receptor (EGFR) is an important target for cancer therapies. Many head and neck cancer (HNC) cells have been reported to overexpress EGFR; therefore, anti-EGFR therapies have been attempted in patients with HNC. However, its clinical efficacy is limited owing to the development of drug resistance. In this study, we developed an EGFR-targeting immunotoxin consisting of a clinically proven anti-EGFR IgG (cetuximab; CTX) and a toxin fragment (LR-LO10) derived from Pseudomonas exotoxin A (PE) using a novel site-specific conjugation technology (peptide-directed photo-crosslinking reaction), as an alternative option. The immunotoxin (CTX-LR-LO10) showed specific binding to EGFR and properties of a typical IgG, such as stability, interactions with receptors of immune cells, and pharmacokinetics, and inhibited protein synthesis via modification of elongation factor-2. Treatment of EGFR-positive HNC cells with the immunotoxin resulted in apoptotic cell death and the inhibition of cell migration and invasion. The efficacy of CTX-LR-LO10 was evaluated in xenograft mouse models, and the immunotoxin exhibited much stronger tumor suppression than CTX or LR-LO10. Transcriptome analyses revealed that the immunotoxins elicited immune responses and altered the expression of genes related to its mechanisms of action. These results support the notion that CTX-LR-LO10 may serve as a new therapeutic agent targeting EGFR-positive cancers.
Collapse
Affiliation(s)
- Mei Huang
- Department of Medical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Jisoo Park
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Jina Seo
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sanghwan Ko
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea
| | - Yoon Hee Yang
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Yeaji Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Hyo Jeong Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Bok-Soon Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yun Sang Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Sciences, Sungshin Women's University, Seoul, Republic of Korea
| | - Sang Teak Jung
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea
- Institute of Human Genetics, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Deachan Park
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, Republic of Korea
| | - Tae Hyeon Yoo
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, Republic of Korea
| | - Chul-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
7
|
Delgado JF, Pritchard WF, Varble N, Lopez-Silva TL, Arrichiello A, Mikhail AS, Morhard R, Ray T, Havakuk MM, Nguyen A, Borde T, Owen JW, Schneider JP, Karanian JW, Wood BJ. X-ray imageable, drug-loaded hydrogel that forms at body temperature for image-guided, needle-based locoregional drug delivery. Sci Rep 2024; 14:13352. [PMID: 38858467 PMCID: PMC11164888 DOI: 10.1038/s41598-024-64189-z] [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: 03/01/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024] Open
Abstract
Liver cancer ranks as the fifth leading cause of cancer-related death globally. Direct intratumoral injections of anti-cancer therapeutics may improve therapeutic efficacy and mitigate adverse effects compared to intravenous injections. Some challenges of intratumoral injections are that the liquid drug formulation may not remain localized and have unpredictable volumetric distribution. Thus, drug delivery varies widely, highly-dependent upon technique. An X-ray imageable poloxamer 407 (POL)-based drug delivery gel was developed and characterized, enabling real-time feedback. Utilizing three needle devices, POL or a control iodinated contrast solution were injected into an ex vivo bovine liver. The 3D distribution was assessed with cone beam computed tomography (CBCT). The 3D distribution of POL gels demonstrated localized spherical morphologies regardless of the injection rate. In addition, the gel 3D conformal distribution could be intentionally altered, depending on the injection technique. When doxorubicin (DOX) was loaded into the POL and injected, DOX distribution on optical imaging matched iodine distribution on CBCT suggesting spatial alignment of DOX and iodine localization in tissue. The controllability and localized deposition of this formulation may ultimately reduce the dependence on operator technique, reduce systemic side effects, and facilitate reproducibility across treatments, through more predictable standardized delivery.
Collapse
Affiliation(s)
- Jose F Delgado
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA.
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
| | - William F Pritchard
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Nicole Varble
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
- Philips Healthcare, Cambridge, MA, USA
| | - Tania L Lopez-Silva
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Center for Cancer Research, Frederick, MD, USA
| | - Antonio Arrichiello
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
- UOS of Interventional Radiology, Department of Diagnostic and Interventional Radiology, Ospedale Maggiore di Lodi, Largo Donatori del Sangue, Lodi, Italy
| | - Andrew S Mikhail
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Robert Morhard
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Trisha Ray
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Michal M Havakuk
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
- Interventional Radiology Department, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Alex Nguyen
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
- Computer Science Department, Stanford University, Stanford, CA, USA
| | - Tabea Borde
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Joshua W Owen
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Joel P Schneider
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Center for Cancer Research, Frederick, MD, USA
| | - John W Karanian
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
| | - Bradford J Wood
- National Institutes of Health, Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, Bethesda, MD, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| |
Collapse
|
8
|
Bertoli E, De Carlo E, Bortolot M, Stanzione B, Del Conte A, Spina M, Bearz A. Targeted Therapy in Mesotheliomas: Uphill All the Way. Cancers (Basel) 2024; 16:1971. [PMID: 38893092 PMCID: PMC11171080 DOI: 10.3390/cancers16111971] [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: 04/06/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Mesothelioma (MM) is an aggressive and lethal disease with few therapeutic opportunities. Platinum-pemetrexed chemotherapy is the backbone of first-line treatment for MM. The introduction of immunotherapy (IO) has been the only novelty of the last decades, allowing an increase in survival compared to standard chemotherapy (CT). However, IO is not approved for epithelioid histology in many countries. Therefore, therapy for relapsed MM remains an unmet clinical need, and the prognosis of MM remains poor, with an average survival of only 18 months. Increasing evidence reveals MM complexity and heterogeneity, of which histological classification fails to explain. Thus, scientific focus on possibly new molecular markers or cellular targets is increasing, together with the search for target therapies directed towards them. The molecular landscape of MM is characterized by inactivating tumor suppressor alterations, the most common of which is found in CDKN2A, BAP1, MTAP, and NF2. In addition, cellular targets such as mesothelin or metabolic enzymes such as ASS1 could be potentially amenable to specific therapies. This review examines the major targets and relative attempts of therapeutic approaches to provide an overview of the potential prospects for treating this rare neoplasm.
Collapse
Affiliation(s)
- Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| |
Collapse
|
9
|
Chakraborty A, Onda M, O’Shea T, Wei J, Liu X, Bera TK, Pastan I. A bispecific antibody that targets the membrane-proximal region of mesothelin and retains high anticancer activity in the presence of shed mesothelin. Mol Cancer Ther 2024; 23:743184. [PMID: 38647528 PMCID: PMC11493849 DOI: 10.1158/1535-7163.mct-23-0233] [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: 04/18/2023] [Revised: 09/03/2023] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Mesothelin (MSLN) is a cell-surface protein that is expressed on many cancers, which makes it a popular target for antibody-based cancer therapy. However, MSLN is shed from cancer cells at high levels via proteases that cleave at its membrane-proximal C-terminal region. Shed MSLN accumulates in patient fluids and tumors and can block antibody-based MSLN-targeting drugs from killing cancer cells. A previously established monoclonal antibody (mAb), 15B6, binds MSLN at its protease-sensitive C-terminal region and does not bind shed MSLN. 15B6 variable fragment (Fv)-derived chimeric antigen receptor (CAR) T cells are not inhibited by shed MSLN and kill tumors in mice more effectively than mAb SS1 Fv-derived CAR T cells, which bind an epitope retained in shed MSLN. Here, we have established 15B6 Fv-derived MSLN x CD3 bispecific antibodies (BsAbs) that target MSLN-expressing cancers. We identified our lead candidate, BsAb 5, after screening multiple 15B6-derived BsAb formats in vitro for cytotoxic activity. BsAb 5 activates T cells to kill various cancer cell lines in a MSLN-specific manner. MSLN 296-591 His, a recombinant protein mimicking shed MSLN, does not inhibit 15B6-derived BsAb 5 but completely inhibits humanized SS1-derived BsAb 7. Furthermore, BsAb 5 inhibits and delays tumor growth and is not inhibited by MSLN 296-585 His in mice. Our findings indicate that by targeting the protease-sensitive region of MSLN, BsAb 5 has high MSLN-specific anticancer activity that is not inhibited by shed MSLN. BsAb 5 may be a promising immunotherapy candidate for MSLN-expressing cancers.
Collapse
Affiliation(s)
- Anirban Chakraborty
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| | - Masanori Onda
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| | - Tara O’Shea
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| | - Junxia Wei
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| | - Xiufen Liu
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| | - Tapan K. Bera
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| | - Ira Pastan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264
| |
Collapse
|
10
|
Skorupan N, Peer CJ, Zhang X, Choo-Wosoba H, Ahmad MI, Lee MJ, Rastogi S, Sato N, Yu Y, Pegna GJ, Steinberg SM, Kalsi SS, Cao L, Figg WD, Trepel JB, Pastan I, FitzGerald D, Alewine C. Tofacitinib to prevent anti-drug antibody formation against LMB-100 immunotoxin in patients with advanced mesothelin-expressing cancers. Front Oncol 2024; 14:1386190. [PMID: 38706610 PMCID: PMC11066227 DOI: 10.3389/fonc.2024.1386190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Background LMB-100 is a mesothelin (MSLN)-targeting recombinant immunotoxin (iTox) carrying a Pseudomonas exotoxin A payload that has shown promise against solid tumors, however, efficacy is limited by the development of neutralizing anti-drug antibodies (ADAs). Tofacitinib is an oral Janus Kinase (JAK) inhibitor that prevented ADA formation against iTox in preclinical studies. Methods A phase 1 trial testing LMB-100 and tofacitinib in patients with MSLN-expressing cancers (pancreatic adenocarcinoma, n=13; cholangiocarcinoma, n=1; appendiceal carcinoma, n=1; cystadenocarcinoma, n=1) was performed to assess safety and to determine if tofacitinib impacted ADA formation. Participants were treated for up to 3 cycles with LMB-100 as a 30-minute infusion on days 4, 6, and 8 at two dose levels (100 and 140 µg/kg) while oral tofacitinib was administered for the first 10 days of the cycle (10 mg BID). Peripheral blood was collected for analysis of ADA levels, serum cytokines and circulating immune subsets. Results The study was closed early due to occurrence of drug-induced pericarditis in 2 patients. Pericarditis with the combination was not reproducible in a transgenic murine model containing human MSLN. Two of 4 patients receiving all 3 cycles of treatment maintained effective LMB-100 levels, an unusual occurrence. Sustained increases in systemic IL-10 and TNF-α were seen, a phenomenon not observed in prior LMB-100 studies. A decrease in activated T cell subsets and an increase in circulating immunosuppressive myeloid populations occurred. No radiologic decreases in tumor volume were observed. Discussion Further testing of tofacitinib to prevent ADA formation is recommended in applicable non-malignant disease settings. Clinical trial registration https://www.clinicaltrials.gov/study/NCT04034238.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xianyu Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mehwish I. Ahmad
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shraddha Rastogi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nahoko Sato
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Guillaume Joe Pegna
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shelley S. Kalsi
- Hematology Consult and Graduate Medical Section, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David FitzGerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christine Alewine
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
11
|
Yun KM, Bazhenova L. Emerging New Targets in Systemic Therapy for Malignant Pleural Mesothelioma. Cancers (Basel) 2024; 16:1252. [PMID: 38610930 PMCID: PMC11011044 DOI: 10.3390/cancers16071252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Malignant pleural mesothelioma (MPM) is a heterogeneous cancer composed of distinct molecular and pathologic subtypes. Unfortunately, MPM is aggressive, and current therapies for advanced, unresectable disease remain limited to cytotoxic chemotherapy and immunotherapy. Our understanding of the genomic landscape of MPM is steadily growing, while the discovery of effective targeted therapies in MPM has advanced more slowly than in other solid tumors. Given the prevalence of alterations in tumor suppressor genes in MPM, it has been challenging to identify actionable targets. However, efforts to characterize the genetic signatures in MPM over the last decade have led to a range of novel targeted therapeutics entering early-phase clinical trials. In this review, we discuss the advancements made thus far in targeted systemic therapies in MPM and the future direction of targeted strategies in patients with advanced MPM.
Collapse
Affiliation(s)
- Karen M. Yun
- Division of Hematology-Oncology, Moores Cancer Center at UC San Diego Health, La Jolla, CA 92093, USA;
| | | |
Collapse
|
12
|
Delgado JF, Pritchard WF, Varble N, Lopez-Silva TL, Arrichiello A, Mikhail AS, Morhard R, Ray T, Havakuk MM, Nguyen A, Borde T, Owen JW, Schneider JP, Karanian JW, Wood BJ. X-ray imageable, drug-loaded hydrogel that forms at body temperature for image-guided, needle- based locoregional drug delivery. RESEARCH SQUARE 2024:rs.3.rs-4003679. [PMID: 38496436 PMCID: PMC10942574 DOI: 10.21203/rs.3.rs-4003679/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Liver cancer ranks as the fifth leading cause of cancer-related death globally. Direct intratumoral injections of anti-cancer therapeutics may improve therapeutic efficacy and mitigate adverse effects compared to intravenous injections. Some challenges of intratumoral injections are that the liquid drug formulation may not remain localized and have unpredictable volumetric distribution. Thus, drug delivery varies widely, highly-dependent upon technique. An x-ray imageable poloxamer 407 (POL)-based drug delivery gel was developed and characterized, enabling real-time feedback. Utilizing three needle devices, POL or a control iodinated contrast solution were injected into an ex vivo bovine liver. The 3D distribution was assessed with cone beam computed tomography (CBCT). The 3D distribution of POL gels demonstrated localized spherical morphologies regardless of the injection rate. In addition, the gel 3D conformal distribution could be intentionally altered, depending on the injection technique. When doxorubicin (DOX) was loaded into the POL and injected, DOX distribution on optical imaging matched iodine distribution on CBCT suggesting spatial alignment of DOX and iodine localization in tissue. The controllability and localized deposition of this formulation may ultimately reduce the dependence on operator technique, reduce systemic side effects, and facilitate reproducibility across treatments, through more predictable standardized delivery.
Collapse
Affiliation(s)
- Jose F Delgado
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - William F Pritchard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | | | - Tania L Lopez-Silva
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Antonio Arrichiello
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Robert Morhard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Trisha Ray
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Michal M Havakuk
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Alex Nguyen
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Tabea Borde
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Joshua W Owen
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Joel P Schneider
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - John W Karanian
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health
| |
Collapse
|
13
|
Ye X, Yu Y, Zheng X, Ma H. Clinical immunotherapy in pancreatic cancer. Cancer Immunol Immunother 2024; 73:64. [PMID: 38430289 PMCID: PMC10908626 DOI: 10.1007/s00262-024-03632-6] [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: 10/07/2023] [Accepted: 01/09/2024] [Indexed: 03/03/2024]
Abstract
Pancreatic cancer remains a challenging disease with limited treatment options, resulting in high mortality rates. The predominant approach to managing pancreatic cancer patients continues to be systemic cytotoxic chemotherapy. Despite substantial advancements in immunotherapy strategies for various cancers, their clinical utility in pancreatic cancer has proven less effective and durable. Whether administered as monotherapy, employing immune checkpoint inhibitors, tumor vaccines, chimeric antigen receptors T cells, or in combination with conventional chemoradiotherapy, the clinical outcomes remain underwhelming. Extensive preclinical experiments and clinical trials in the realm of pancreatic cancer have provided valuable insights into the complexities of immunotherapy. Chief among the hurdles are the immunosuppressive tumor microenvironment, limited immunogenicity, and the inherent heterogeneity of pancreatic cancer. In this comprehensive review, we provide an overview and critical analysis of current clinical immunotherapy strategies for pancreatic cancer, emphasizing their endeavors to overcome immunotherapy resistance. Particular focus is placed on strategies aimed at reshaping the immunosuppressive microenvironment and enhancing T cell-mediated tumor cell killing. Ultimately, through deeper elucidation of the underlying pathogenic mechanisms of pancreatic cancer and the refinement of therapeutic approaches, we anticipate breakthroughs that will pave the way for more effective treatments in this challenging disease.
Collapse
Affiliation(s)
- Xiaorong Ye
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China
| | - Yue Yu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China.
| | - Xiaohu Zheng
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China.
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China.
| | - Hongdi Ma
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.
- Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui Province, People's Republic of China.
| |
Collapse
|
14
|
Schaffrath R, Brinkmann U. Diphthamide - a conserved modification of eEF2 with clinical relevance. Trends Mol Med 2024; 30:164-177. [PMID: 38097404 DOI: 10.1016/j.molmed.2023.11.008] [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: 09/28/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 02/17/2024]
Abstract
Diphthamide, a complex modification on eukaryotic translation elongation factor 2 (eEF2), assures reading-frame fidelity during translation. Diphthamide and enzymes for its synthesis are conserved in eukaryotes and archaea. Originally identified as target for diphtheria toxin (DT) in humans, its clinical relevance now proves to be broader than the link to pathogenic bacteria. Diphthamide synthesis enzymes (DPH1 and DPH3) are associated with cancer, and DPH gene mutations can cause diphthamide deficiency syndrome (DDS). Finally, new analyses provide evidence that diphthamide may restrict propagation of viruses including SARS-CoV-2 and HIV-1, and that DPH enzymes are targeted by viruses for degradation to overcome this restriction. This review describes how diphthamide is synthesized and functions in translation, and covers its clinical relevance in human development, cancer, and infectious diseases.
Collapse
Affiliation(s)
- Raffael Schaffrath
- Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, Kassel, Germany.
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany.
| |
Collapse
|
15
|
Nijhawan HP, Prabhakar B, Misra A, Yadav KS. Fragmented antibodies in non-small cell lung cancer: A novel nano-engineered delivery system for detection and treatment of cancer. Drug Discov Today 2023; 28:103701. [PMID: 37453459 DOI: 10.1016/j.drudis.2023.103701] [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: 09/03/2022] [Revised: 05/08/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Non-small cell lung cancer (NSCLC) has a long history of defying traditional cytotoxic treatment. Significant advancements in biotechnology, cancer biology, and immunotherapy have provided new insights that have altered the landscape for the management of NSCLC, clearing the way for a new era of pharmaceuticals in the form of monoclonal antibodies and their fragments. Antibody fragments are superior to monoclonal antibodies because of their small size, which allows them to penetrate cells and tissues effectively. When combined with functional nanocarriers, antibody fragments can target cancer cells while offering improved efficacy and fewer off-target effects. We discuss current topics of interest including anti-CTLA-4 mAbs, Talactoferrin alfa (TLF), and the CYFRA 21-1 biomarker, with brief insights into its novel detection system.
Collapse
Affiliation(s)
- Harsh P Nijhawan
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India
| | - Bala Prabhakar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India
| | - Ambikanandan Misra
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India.
| |
Collapse
|
16
|
Qualiotto AN, Baldavira CM, Balancin M, Ab’Saber A, Takagaki T, Capelozzi VL. Mesothelin expression remodeled the immune-matrix tumor microenvironment predicting the risk of death in patients with malignant pleural mesothelioma. Front Immunol 2023; 14:1268927. [PMID: 37901248 PMCID: PMC10601658 DOI: 10.3389/fimmu.2023.1268927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Background The combination of immunobiological agents with immune checkpoint proteins is a promising treatment for malignant pleural mesothelioma (MPM). Mesothelin and anti-PD-L1 antibody-drug conjugates specifically target malignant neoplastic cells, inhibit the migration and invasion of neoplastic cells, and restore the immune landscape. In this study, we confirmed the importance of mesothelin and examined the relationship between mesothelin and the immune landscape of the tumor microenvironment (TME) in two MPM cohorts. Methods The discovery cohort included 82 MPM cases. Tissue microarray slides were generated, and samples were processed for hematoxylin & eosin staining, immunohistochemistry, and immunofluorescence assays. The relationship between mesothelin, biomarkers of histogenesis, histological aggressiveness, PD-L1, immune cells (CD4, CD8, CD20, CD68), and collagen type I and type V fibers was evaluated by quantitative digital analyses. The outcome was the survival time until death from disease recurrence. The exploratory cohort included 87 malignant mesothelioma (MESO) patients from The Cancer Genome Atlas database. Results Most patients were male (70.7%) with a history of asbestos exposure (53.7%) and with the epithelioid subtype (89%). Surgical resection was performed in 85.4% of patients, and 14.6% received chemotherapy; 59.8% of patients died from disease extension to the mediastinum. Low tumor mesothelin expression was associated with tumor necrosis and nuclear grade 1, whereas high mesothelin expression was significantly associated with the epithelioid histotype and high density of T cells CD8+, macrophages CD68+, and collagen type I fibers. Cox multivariate analysis showed a high risk of death for non-operated patients [hazard ratio (HR), 3.42 (1.15-10.16)] with low tumor mesothelin levels [HR, 2.58 (1.09-6.10)] and high PD-L1 and low infiltration of T cells CD4+ [HR, 3.81 (1.58-9.18)]. In the exploratory cohort, low mesothelin and high COL1A1 and COL5A1 expression were associated with poor overall survival. Conclusion Tumor mesothelin expression associated with the TME immune landscape predicts the risk of death for patients with MPM and could be a new target for immunotherapy in MPM.
Collapse
Affiliation(s)
- Aline Nery Qualiotto
- Laboratory of Genomic and Histomorphometry, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Camila Machado Baldavira
- Laboratory of Genomic and Histomorphometry, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Marcelo Balancin
- Laboratory of Genomic and Histomorphometry, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Alexandre Ab’Saber
- Laboratory of Genomic and Histomorphometry, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Teresa Takagaki
- Division of Pneumology, Heart Institute (Incor), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Laboratory of Genomic and Histomorphometry, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
17
|
Wang Q, Xu C, Wang W, Zhang Y, Li Z, Song Z, Wang J, Yu J, Liu J, Zhang S, Cai X, Li W, Zhan P, Liu H, Lv T, Miao L, Min L, Li J, Liu B, Yuan J, Jiang Z, Lin G, Chen X, Pu X, Rao C, Lv D, Yu Z, Li X, Tang C, Zhou C, Zhang J, Guo H, Chu Q, Meng R, Liu X, Wu J, Hu X, Zhou J, Zhu Z, Chen X, Pan W, Pang F, Zhang W, Jian Q, Wang K, Wang L, Zhu Y, Yang G, Lin X, Cai J, Feng H, Wang L, Du Y, Yao W, Shi X, Niu X, Yuan D, Yao Y, Huang J, Wang X, Zhang Y, Sun P, Wang H, Ye M, Wang D, Wang Z, Hao Y, Wang Z, Wan B, Lv D, Yu J, Kang J, Zhang J, Zhang C, Wu L, Shi L, Ye L, Wang G, Wang Y, Gao F, Huang J, Wang G, Wei J, Huang L, Li B, Zhang Z, Li Z, Liu Y, Li Y, Liu Z, Yang N, Wu L, Wang Q, Huang W, Hong Z, Wang G, Qu F, Fang M, Fang Y, Zhu X, et alWang Q, Xu C, Wang W, Zhang Y, Li Z, Song Z, Wang J, Yu J, Liu J, Zhang S, Cai X, Li W, Zhan P, Liu H, Lv T, Miao L, Min L, Li J, Liu B, Yuan J, Jiang Z, Lin G, Chen X, Pu X, Rao C, Lv D, Yu Z, Li X, Tang C, Zhou C, Zhang J, Guo H, Chu Q, Meng R, Liu X, Wu J, Hu X, Zhou J, Zhu Z, Chen X, Pan W, Pang F, Zhang W, Jian Q, Wang K, Wang L, Zhu Y, Yang G, Lin X, Cai J, Feng H, Wang L, Du Y, Yao W, Shi X, Niu X, Yuan D, Yao Y, Huang J, Wang X, Zhang Y, Sun P, Wang H, Ye M, Wang D, Wang Z, Hao Y, Wang Z, Wan B, Lv D, Yu J, Kang J, Zhang J, Zhang C, Wu L, Shi L, Ye L, Wang G, Wang Y, Gao F, Huang J, Wang G, Wei J, Huang L, Li B, Zhang Z, Li Z, Liu Y, Li Y, Liu Z, Yang N, Wu L, Wang Q, Huang W, Hong Z, Wang G, Qu F, Fang M, Fang Y, Zhu X, Du K, Ji J, Shen Y, Chen J, Zhang Y, Ma S, Lu Y, Song Y, Liu A, Zhong W, Fang W. Chinese expert consensus on the diagnosis and treatment of malignant pleural mesothelioma. Thorac Cancer 2023; 14:2715-2731. [PMID: 37461124 PMCID: PMC10493492 DOI: 10.1111/1759-7714.15022] [Show More Authors] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 09/12/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a malignant tumor originating from the pleura, and its incidence has been increasing in recent years. Due to the insidious onset and strong local invasiveness of MPM, most patients are diagnosed in the late stage and early screening and treatment for high-risk populations are crucial. The treatment of MPM mainly includes surgery, chemotherapy, and radiotherapy. Immunotherapy and electric field therapy have also been applied, leading to further improvements in patient survival. The Mesothelioma Group of the Yangtze River Delta Lung Cancer Cooperation Group (East China LUng caNcer Group, ECLUNG; Youth Committee) developed a national consensus on the clinical diagnosis and treatment of MPM based on existing clinical research evidence and the opinions of national experts. This consensus aims to promote the homogenization and standardization of MPM diagnosis and treatment in China, covering epidemiology, diagnosis, treatment, and follow-up.
Collapse
Affiliation(s)
- Qian Wang
- Department of Respiratory MedicineAffiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Chunwei Xu
- Institute of Cancer and Basic Medicine (ICBM)Chinese Academy of SciencesHangzhouChina
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Wenxian Wang
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Ziming Li
- Department of Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Zhengbo Song
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Jiandong Wang
- Department of PathologyAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Jinpu Yu
- Department of Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Jingjing Liu
- Department of Thoracic CancerJilin Cancer HospitalChangchunChina
| | - Shirong Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer CenterZhejiang University School of MedicineHangzhouChina
| | - Xiuyu Cai
- Department of VIP Inpatient, Sun Yet‐Sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Cancer CenterZhejiang UniversityHangzhouChina
| | - Ping Zhan
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Hongbing Liu
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Tangfeng Lv
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Liyun Miao
- Department of Respiratory Medicine, Affiliated Drum Tower HospitalMedical School of Nanjing UniversityNanjingChina
| | - Lingfeng Min
- Department of Respiratory MedicineClinical Medical School of Yangzhou University, Subei People's Hospital of Jiangsu ProvinceYangzhouChina
| | - Jiancheng Li
- Department of Radiation OncologyFujian Medical University Cancer Hospital & Fujian Cancer HospitalFuzhouChina
| | - Baogang Liu
- Department of OncologyHarbin Medical University Cancer HospitalHarbinChina
| | - Jingping Yuan
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhansheng Jiang
- Department of Integrative OncologyTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Gen Lin
- Department of Medical OncologyFujian Medical University Cancer Hospital & Fujian Cancer HospitalFuzhouChina
| | - Xiaohui Chen
- Department of Thoracic SurgeryFujian Medical University Cancer Hospital & Fujian Cancer HospitalFuzhouChina
| | - Xingxiang Pu
- Department of Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Chuangzhou Rao
- Department of Radiotherapy and Chemotherapy, Hwamei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Dongqing Lv
- Department of Pulmonary MedicineTaizhou Hospital of Wenzhou Medical UniversityTaizhouChina
| | - Zongyang Yu
- Department of Respiratory Medicine, the 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital)Fujian Medical UniversityFuzhouChina
| | - Xiaoyan Li
- Department of Oncology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Chuanhao Tang
- Department of Medical OncologyPeking University International HospitalBeijingChina
| | - Chengzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University(The First Affiliated Hospital of Guangzhou Medical University)GuangzhouChina
| | - Junping Zhang
- Department of Thoracic OncologyShanxi Academy of Medical Sciences, Shanxi Bethune HospitalTaiyuanChina
| | - Hui Guo
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuewen Liu
- Department of Oncology, the Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Jingxun Wu
- Department of Medical Oncology, the First Affiliated Hospital of MedicineXiamen UniversityXiamenChina
| | - Xiao Hu
- Zhejiang Key Laboratory of Radiation OncologyCancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)HangzhouChina
| | - Jin Zhou
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and TechnologyChengduChina
| | - Zhengfei Zhu
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Xiaofeng Chen
- Department of OncologyJiangsu Province Hospital and Nanjing Medical University First Affiliated HospitalNanjingChina
| | - Weiwei Pan
- Department of Cell Biology, College of MedicineJiaxing UniversityJiaxingChina
| | - Fei Pang
- Department of MedicalShanghai OrigiMed Co, LtdShanghaiChina
| | - Wenpan Zhang
- Department of MedicalShanghai OrigiMed Co, LtdShanghaiChina
| | - Qijie Jian
- Department of MedicalShanghai OrigiMed Co, LtdShanghaiChina
| | - Kai Wang
- Department of MedicalShanghai OrigiMed Co, LtdShanghaiChina
| | - Liping Wang
- Department of OncologyBaotou Cancer HospitalBaotouChina
| | - Youcai Zhu
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun HospitalThe Third Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Guocai Yang
- Department of Thoracic Surgery, Zhoushan HospitalWenzhou Medical UniversityZhoushanChina
| | - Xinqing Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University(The First Affiliated Hospital of Guangzhou Medical University)GuangzhouChina
| | - Jing Cai
- Department of OncologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Huijing Feng
- Department of Thoracic OncologyShanxi Academy of Medical Sciences, Shanxi Bethune HospitalTaiyuanChina
| | - Lin Wang
- Department of PathologyShanxi Academy of Medical Sciences, Shanxi Bethune HospitalTaiyuanChina
| | - Yingying Du
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Wang Yao
- Department of Interventional OncologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Xuefei Shi
- Department of Respiratory Medicine, Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Dongmei Yuan
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Yanwen Yao
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Jianhui Huang
- Department of OncologyLishui Municipal Central HospitalLishuiChina
| | - Xiaomin Wang
- Department of Cell Biology, College of MedicineJiaxing UniversityJiaxingChina
| | - Yinbin Zhang
- Department of Oncologythe Second Affiliated Hospital of Medical College, Xi'an Jiaotong UniversityXi'anChina
| | - Pingli Sun
- Department of PathologyThe Second Hospital of Jilin UniversityChangchunChina
| | - Hong Wang
- Senior Department of OncologyThe 5th Medical Center of PLA General HospitalBeijingChina
| | - Mingxiang Ye
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Dong Wang
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Zhaofeng Wang
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Yue Hao
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Zhen Wang
- Department of Radiation OncologyAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Bing Wan
- Department of Respiratory MedicineThe Affiliated Jiangning Hospital of Nanjing Medical UniversityNanjingChina
| | - Donglai Lv
- Department of Clinical OncologyThe 901 Hospital of Joint Logistics Support Force of People Liberation ArmyHefeiChina
| | - Jianwei Yu
- Department of Respiratory MedicineAffiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi Province Hospital of Chinese MedicineNanchangChina
| | - Jin Kang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung CancerGuangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of MedicineGuangzhouChina
| | - Jiatao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung CancerGuangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of MedicineGuangzhouChina
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung CancerGuangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of MedicineGuangzhouChina
| | - Lixin Wu
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun HospitalThe Third Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Lin Shi
- Department of Respiratory MedicineZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Leiguang Ye
- Department of OncologyHarbin Medical University Cancer HospitalHarbinChina
| | - Gaoming Wang
- Department of Thoracic Surgery, Xuzhou Central HospitalXuzhou Clinical School of Xuzhou Medical UniversityXuzhouChina
| | - Yina Wang
- Department of Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Feng Gao
- Department of Thoracic SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Jianfei Huang
- Department of Clinical BiobankAffiliated Hospital of Nantong UniversityNantongChina
| | - Guifang Wang
- Department of Respiratory MedicineHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jianguo Wei
- Department of PathologyShaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)ShaoxingChina
| | - Long Huang
- Department of OncologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Bihui Li
- Department of OncologyThe Second Affiliated Hospital of Guilin Medical UniversityGuilinChina
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of PharmacyJinan UniversityGuangzhouChina
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of PathologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yuan Li
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Zhefeng Liu
- Senior Department of OncologyThe 5th Medical Center of PLA General HospitalBeijingChina
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Lin Wu
- Department of Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Qiming Wang
- Department of Internal MedicineThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Wenbin Huang
- Department of Pathologythe First Affiliated Hospital of Henan University of Science and TechnologyLuoyangChina
| | - Zhuan Hong
- Department of Medical Oncology, Jiangsu Cancer HospitalNanjing Medical University Affiliated Cancer HospitalNanjingChina
| | - Guansong Wang
- Institute of Respiratory Diseases, Xinjian HospitalThird Military Medical UniversityChongqingChina
| | - Fengli Qu
- Institute of Cancer and Basic Medicine (ICBM)Chinese Academy of SciencesHangzhouChina
| | - Meiyu Fang
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang UniversityHangzhouChina
| | - Xixu Zhu
- Department of Radiation OncologyAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Kaiqi Du
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun HospitalThe Third Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Jiansong Ji
- Department of RadiologyLishui Municipal Central HospitalLishuiChina
| | - Yi Shen
- Department of Thoracic Surgery, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yiping Zhang
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Shenglin Ma
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Cancer CenterZhejiang University School of MedicineHangzhouChina
| | - Yuanzhi Lu
- Department of Clinical PathologyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yong Song
- Department of Respiratory MedicineAffiliated Jinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Anwen Liu
- Department of OncologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung CancerGuangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of MedicineGuangzhouChina
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| |
Collapse
|
18
|
Dagogo-Jack I. Targeted Approaches to Treatment of Pleural Mesothelioma: A Review. JCO Precis Oncol 2023; 7:e2300344. [PMID: 37992257 PMCID: PMC10681489 DOI: 10.1200/po.23.00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 11/24/2023] Open
Abstract
Pleural mesothelioma is an aggressive disease that is enriched for inactivating alterations in tumor suppressor genes. Systemic therapeutic strategies for pleural mesothelioma generally involve chemotherapies and immunotherapies that are chosen without consideration of the tumor's molecular profile. As this generalized approach to treatment rarely yields durable responses, alternative therapeutic regimens are urgently indicated. Preclinical studies have identified synthetic lethal protein and metabolic interactions, recurrently overexpressed proteins, and frequent pathway perturbations that may be therapeutically exploited in mesothelioma. This review discusses the mechanism of action of emerging investigational therapies and summarizes findings from phase I-II clinical trials exploring selective, biomarker-driven therapeutic strategies for mesothelioma, with a focus on five common targets. Finally, using lessons learned from these clinical trials, imperatives for successful implementation of targeted therapy in mesothelioma are discussed.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
19
|
Hassan R, Butler M, O'Cearbhaill RE, Oh DY, Johnson M, Zikaras K, Smalley M, Ross M, Tanyi JL, Ghafoor A, Shah NN, Saboury B, Cao L, Quintás-Cardama A, Hong D. Mesothelin-targeting T cell receptor fusion construct cell therapy in refractory solid tumors: phase 1/2 trial interim results. Nat Med 2023; 29:2099-2109. [PMID: 37501016 PMCID: PMC10427427 DOI: 10.1038/s41591-023-02452-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/08/2023] [Indexed: 07/29/2023]
Abstract
The T cell receptor fusion construct (TRuC) gavocabtagene autoleucel (gavo-cel) consists of single-domain anti-mesothelin antibody that integrates into the endogenous T cell receptor (TCR) and engages the signaling capacity of the entire TCR upon mesothelin binding. Here we describe phase 1 results from an ongoing phase1/2 trial of gavo-cel in patients with treatment-refractory mesothelin-expressing solid tumors. The primary objectives were to evaluate safety and determine the recommended phase 2 dose (RP2D). Secondary objectives included efficacy. Thirty-two patients received gavo-cel at increasing doses either as a single agent (n = 3) or after lymphodepletion (LD, n = 29). Dose-limiting toxicities of grade 3 pneumonitis and grade 5 bronchioalveolar hemorrhage were noted. The RP2D was determined as 1 × 108 cells per m2 after LD. Grade 3 or higher pneumonitis was seen in 16% of all patients and in none at the RP2D; grade 3 or higher cytokine release syndrome occurred in 25% of all patients and in 15% at the RP2D. In 30 evaluable patients, the overall response rate and disease control rate were 20% (13% confirmed) and 77%, respectively, and the 6-month overall survival rate was 70%. Gavo-cel warrants further study in patients with mesothelin-expressing cancers given its encouraging anti-tumor activity, but it may have a narrow therapeutic window. ClinicalTrials.gov identifier: NCT03907852 .
Collapse
Affiliation(s)
- Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Marcus Butler
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Roisin E O'Cearbhaill
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | | | - Janos L Tanyi
- Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA, USA
| | - Azam Ghafoor
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
20
|
Ray A, Moore TF, Pandit R, Burke AD, Borsch DM. An Overview of Selected Bacterial Infections in Cancer, Their Virulence Factors, and Some Aspects of Infection Management. BIOLOGY 2023; 12:963. [PMID: 37508393 PMCID: PMC10376897 DOI: 10.3390/biology12070963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/30/2023] [Accepted: 07/02/2023] [Indexed: 07/30/2023]
Abstract
In cancer development and its clinical course, bacteria can be involved in etiology and secondary infection. Regarding etiology, various epidemiological studies have revealed that Helicobacter pylori can directly impact gastric carcinogenesis. The Helicobacter pylori-associated virulence factor cytotoxin-associated gene A perhaps plays an important role through different mechanisms such as aberrant DNA methylation, activation of nuclear factor kappa B, and modulation of the Wnt/β-catenin signaling pathway. Many other bacteria, including Salmonella and Pseudomonas, can also affect Wnt/β-catenin signaling. Although Helicobacter pylori is involved in both gastric adenocarcinoma and mucosa-associated lymphoid tissue lymphoma, its role in the latter disease is more complicated. Among other bacterial species, Chlamydia is linked with a diverse range of diseases including cancers of different sites. The cellular organizations of Chlamydia are highly complex. Interestingly, Escherichia coli is believed to be associated with colon cancer development. Microorganisms such as Escherichia coli and Pseudomonas aeruginosa are frequently isolated from secondary infections in cancer patients. In these patients, the common sites of infection are the respiratory, gastrointestinal, and urinary tracts. There is an alarming rise in infections with multidrug-resistant bacteria and the scarcity of suitable antimicrobial agents adversely influences prognosis. Therefore, effective implementation of antimicrobial stewardship strategies is important in cancer patients.
Collapse
Affiliation(s)
- Amitabha Ray
- College of Medical Science, Alderson Broaddus University, 101 College Hill Drive, Philippi, WV 26416, USA
| | - Thomas F Moore
- College of Medical Science, Alderson Broaddus University, 101 College Hill Drive, Philippi, WV 26416, USA
| | | | | | - Daniel M Borsch
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA 15601, USA
| |
Collapse
|
21
|
Offin M, Fitzgerald B, Zauderer MG, Doroshow D. The past, present, and future of targeted therapeutic approaches in patients with diffuse pleural mesotheliomas. JOURNAL OF CANCER METASTASIS AND TREATMENT 2023; 9:21. [PMID: 38895597 PMCID: PMC11185317 DOI: 10.20517/2394-4722.2022.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Despite our growing understanding of the genomic landscape of diffuse pleural mesotheliomas (DPM), there has been limited success in targeted therapeutic strategies for the disease. This review summarizes attempts to develop targeted therapies in DPM, focusing on the following targets being clinically explored in recent and ongoing clinical trials: vascular endothelial growth factor, mesothelin, BRCA1-associated protein 1, Wilms tumor 1 protein, NF2/YAP/TAZ, CDKN2, methylthioadenosine phosphorylase, v-domain Ig suppressor T-cell activation, and argininosuccinate synthetase 1. Although preclinical data for these targets are promising, few have efficaciously translated to benefit our patients. Future efforts should seek to expand the availability of preclinical models that faithfully recapitulate DPM biology, develop clinically relevant biomarkers, and refine patient selection criteria for clinical trials.
Collapse
Affiliation(s)
- Michael Offin
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bailey Fitzgerald
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marjorie G. Zauderer
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY 10065, USA
| | - Deborah Doroshow
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
22
|
Borea F, Franczak MA, Garcia M, Perrino M, Cordua N, Smolenski RT, Peters GJ, Dziadziuszko R, Santoro A, Zucali PA, Giovannetti E. Target Therapy in Malignant Pleural Mesothelioma: Hope or Mirage? Int J Mol Sci 2023; 24:9165. [PMID: 37298116 PMCID: PMC10253134 DOI: 10.3390/ijms24119165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
Malignant Pleural Mesothelioma (MPM) is a rare neoplasm that is typically diagnosed in a locally advanced stage, making it not eligible for radical surgery and requiring systemic treatment. Chemotherapy with platinum compounds and pemetrexed has been the only approved standard of care for approximately 20 years, without any relevant therapeutic advance until the introduction of immune checkpoint inhibitors. Nevertheless, the prognosis remains poor, with an average survival of only 18 months. Thanks to a better understanding of the molecular mechanisms underlying tumor biology, targeted therapy has become an essential therapeutic option in several solid malignancies. Unfortunately, most of the clinical trials evaluating potentially targeted drugs for MPM have failed. This review aims to present the main findings of the most promising targeted therapies in MPM, and to explore possible reasons leading to treatments failures. The ultimate goal is to determine whether there is still a place for continued preclinical/clinical research in this area.
Collapse
Affiliation(s)
- Federica Borea
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Department of Medical Oncology, Amsterdam University Medical Centers, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marika A. Franczak
- Department of Medical Oncology, Amsterdam University Medical Centers, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdańsk, Poland
| | - Maria Garcia
- Faculty of Experimental Science, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Matteo Perrino
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Nadia Cordua
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | | | - Godefridus J. Peters
- Department of Medical Oncology, Amsterdam University Medical Centers, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdańsk, Poland
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy and Early Phase Clinical Trials Centre, Medical University of Gdansk, 80-210 Gdańsk, Poland
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Paolo A. Zucali
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Centers, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Fondazione Pisana per la Scienza, 56017 Pisa, Italy
| |
Collapse
|
23
|
Targeting Mesothelin in Solid Tumours: Anti-mesothelin Antibody and Drug Conjugates. Curr Oncol Rep 2023; 25:309-323. [PMID: 36763234 DOI: 10.1007/s11912-023-01367-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 02/11/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarise the pathobiological role of mesothelin and the current data on therapeutic antibodies targeting mesothelin in solid tumours. RECENT FINDINGS High mesothelin expression is restricted to the pericardium, pleura, peritoneum and tunica vaginalis. Mesothelin does not seem to have any normal biological function in adult normal tissues. Mesothelin is highly expressed in mesothelioma, serous ovarian cancer, pancreatic cancer and some gastric cancer and adenocarcinoma of the lung and is responsible for tumour proliferation, metastasis, resistance to chemotherapy or radiation and evasion of immune system. To date, antibody, antibody drug conjugates and bispecific antibodies with immune checkpoints have been investigated in mesothelin expressing malignancies. After a couple of decades of clinical investigation in antibody targeting mesothelin, the therapeutic benefit is relatively modest. Novel delivery of mesothelin targeting agents, more potent payload in antibody drug conjugates and immune checkpoint inhibitor, may improve therapeutic benefit.
Collapse
|
24
|
Pegna GJ, Lee M, Peer CJ, Ahmad MI, Venzon DJ, Yu Y, Yuno A, Steinberg SM, Cao L, Figg WD, Donahue RN, Hassan R, Pastan I, Trepel JB, Alewine C. Systemic immune changes accompany combination treatment with immunotoxin LMB-100 and nab-paclitaxel. Cancer Med 2023; 12:4236-4249. [PMID: 36208017 PMCID: PMC9972172 DOI: 10.1002/cam4.5290] [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: 03/23/2022] [Revised: 07/20/2022] [Accepted: 09/12/2022] [Indexed: 11/07/2022] Open
Abstract
LMB-100 is a novel immune-conjugate (immunotoxin) that targets mesothelin. A phase 1/2 clinical trial was conducted (NCT02810418) with primary objectives assessing the safety and efficacy of LMB-100 ± nab-paclitaxel. Participant blood samples were analyzed for changes in serum cytokines and circulating immune cell subsets associated with response or toxicity. On Arm A, participants (n = 20) received standard 30-minute LMB-100 infusion with nab-paclitaxel. Although clinical efficacy was observed, the combination caused intolerable capillary leak syndrome (CLS), a major toxicity of unclear etiology that affects many immunotoxin drugs. Participants developing CLS experienced rapid elevations in IFNγ and IL-8 compared to those without significant CLS, along with midcycle increases in Ki-67- CD4 T cells that were CD38, HLA-DR, or TIM3 positive. Additionally, a strong increase in activated CD4 and CD8 T cells and a concurrent decrease in Tregs were seen in the single Arm A patient achieving a partial response. In Arm B, administration of single agent LMB-100 to participants (n = 20) as a long infusion given over 24-48 h was investigated based on pre-clinical data that this format could reduce CLS. An optimal dose and schedule of long infusion LMB-100 were identified, but no clinical efficacy was observed even in patients receiving LMB-100 in combination with nab-paclitaxel. Despite this, both Arm A and B participants experienced increases in specific subsets of proliferating CD4 and CD8 T cells following Cycle 1 treatment. In summary, LMB-100 treatment causes systemic immune activation. Inflammatory and immune changes that accompany drug associated CLS were characterized for the first time.
Collapse
Affiliation(s)
- Guillaume Joe Pegna
- Laboratory of Molecular BiologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Medical Oncology ProgramNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Knight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Min‐Jung Lee
- Developmental Therapeutics BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Cody J. Peer
- Clinical Pharmacology ProgramNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Mehwish I. Ahmad
- Office of Research NursingNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Astra ZenecaGaithersburgMarylandUSA
| | - David J. Venzon
- Biostatistics and Data Management SectionNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Yunkai Yu
- Genetics BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Akira Yuno
- Developmental Therapeutics BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Oral and Maxillofacial SurgeryKumamoto University HospitalKumamotoJapan
| | - Seth M. Steinberg
- Biostatistics and Data Management SectionNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Liang Cao
- Genetics BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - William D. Figg
- Clinical Pharmacology ProgramNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Renee N. Donahue
- Laboratory of Tumor Immunology and BiologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Raffit Hassan
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Ira Pastan
- Laboratory of Molecular BiologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Jane B. Trepel
- Developmental Therapeutics BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Christine Alewine
- Laboratory of Molecular BiologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
25
|
Takamizawa S, Yazaki S, Kojima Y, Yoshida H, Kitadai R, Nishikawa T, Shimoi T, Sudo K, Okuma HS, Tanioka M, Noguchi E, Uno M, Ishikawa M, Kato T, Fujiwara Y, Yonemori K. High mesothelin expression is correlated with non-squamous cell histology and poor survival in cervical cancer: a retrospective study. BMC Cancer 2022; 22:1215. [PMID: 36434635 PMCID: PMC9701073 DOI: 10.1186/s12885-022-10277-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Mesothelin (MSLN) is a cell-surface glycoprotein found in various solid tumours. Cancer therapies targeting MSLN have been developed in recent years; however, the available information on MSLN expression in cervical cancer is limited. This study aimed to evaluate MSLN expression in various histological types of cervical cancer and examine its relationship with prognosis. METHODS This retrospective study included patients with cervical cancer who underwent primary surgery between January 2000 and December 2020 at our institution. MSLN expression was evaluated by immunohistochemistry using clone SP74 and defined as positive if MSLN was expressed at any intensity. High MSLN expression was defined as an intensity of ≥ 2 + in ≥ 30% of tumour cells. The association between MSLN expression and clinicopathological factors was evaluated. RESULTS Overall, 123 patients were identified, and 140 tumour samples, including 17 paired primary and metastatic samples, were evaluated. Concerning histological type, 67 patients had squamous cell carcinoma (SCC), whereas 56 had non-SCC. MSLN expression was observed in 98.4% (121/123) of primary tumours. High MSLN expression was observed in 63.4% of samples (78/123), but it differed between the histological types (49.2% for SCC vs. 80.4% for non-SCC, p < 0.001). There was a significant correlation between MSLN expression in primary and metastatic lesions (Rs = 0.557, p = 0.015). In patients with common histological types, overall survival (OS) was shorter in the high MSLN expression group than in the low MSLN expression group (hazard ratio, 3.53; 95% confidence interval, 1.16-15.3, p = 0.03). CONCLUSIONS MSLN was highly expressed in patients with cervical cancer, especially in those with non-SCC. High MSLN expression in the primary lesion was significantly associated with poor OS, and its expression was maintained in metastatic lesions. Our findings indicate that MSLN may be an attractive therapeutic target for cervical cancer. TRIAL REGISTRATION Retrospectively registered. 2014-393. 1 June 2015.
Collapse
Affiliation(s)
- Shigemasa Takamizawa
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Shu Yazaki
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Yuki Kojima
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Hiroshi Yoshida
- grid.272242.30000 0001 2168 5385Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Rui Kitadai
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Tadaaki Nishikawa
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Tatsunori Shimoi
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Kazuki Sudo
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Hitomi Sumiyoshi Okuma
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Maki Tanioka
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Emi Noguchi
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Masaya Uno
- grid.272242.30000 0001 2168 5385Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Mitsuya Ishikawa
- grid.272242.30000 0001 2168 5385Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Tomoyasu Kato
- grid.272242.30000 0001 2168 5385Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuhiro Fujiwara
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Kan Yonemori
- grid.272242.30000 0001 2168 5385Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| |
Collapse
|
26
|
Nishio T, Koyama Y, Fuji H, Ishizuka K, Iwaisako K, Taura K, Hatano E, Brenner DA, Kisseleva T. The Role of Mesothelin in Activation of Portal Fibroblasts in Cholestatic Liver Injury. BIOLOGY 2022; 11:1589. [PMID: 36358290 PMCID: PMC9687690 DOI: 10.3390/biology11111589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022]
Abstract
Fibrosis is a common consequence of abnormal wound healing, which is characterized by infiltration of myofibroblasts and formation of fibrous scar. In liver fibrosis, activated Hepatic Stellate Cells (aHSCs) and activated Portal Fibroblasts (aPFs) are the major contributors to the origin of hepatic myofibroblasts. aPFs are significantly involved in the pathogenesis of cholestatic fibrosis, suggesting that aPFs may be a primary target for anti-fibrotic therapy in cholestatic injury. aPFs are distinguishable from aHSCs by specific markers including mesothelin (Msln), Mucin 16 (Muc16), and Thymus cell antigen 1 (Thy1, CD90) as well as fibulin 2, elastin, Gremlin 1, ecto-ATPase nucleoside triphosphate diphosphohydrolase 2. Msln plays a critical role in activation of PFs, via formation of Msln-Muc16-Thy1 complex that regulates TGFβ1/TGFβRI-mediated fibrogenic signaling. The opposing pro- and anti-fibrogenic effects of Msln and Thy1 are key components of the TGFβ1-induced activation pathway in aPFs. In addition, aPFs and activated lung and kidney fibroblasts share similarities across different organs with expression of common markers and activation cascade including Msln-Thy1 interaction. Here, we summarize the potential function of Msln in activation of PFs and development of cholestatic fibrosis, offering a novel perspective for anti-fibrotic therapy targeting Msln.
Collapse
Affiliation(s)
- Takahiro Nishio
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yukinori Koyama
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroaki Fuji
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| | - Kei Ishizuka
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| | - Keiko Iwaisako
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe 610-0394, Japan
| | - Kojiro Taura
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Gastroenterological Surgery and Oncology, Kitano Hospital Medical Research Institute, 2-4-20 Ogimachi, Kita-ku, Osaka 530-8480, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - David A. Brenner
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, 9500 Gilman Drive, #0063, La Jolla, CA 92093, USA
| |
Collapse
|
27
|
Dicks LMT, Vermeulen W. Do Bacteria Provide an Alternative to Cancer Treatment and What Role Does Lactic Acid Bacteria Play? Microorganisms 2022; 10:microorganisms10091733. [PMID: 36144335 PMCID: PMC9501580 DOI: 10.3390/microorganisms10091733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide. According to 2022 statistics from the World Health Organization (WHO), close to 10 million deaths have been reported in 2020 and it is estimated that the number of cancer cases world-wide could increase to 21.6 million by 2030. Breast, lung, thyroid, pancreatic, liver, prostate, bladder, kidney, pelvis, colon, and rectum cancers are the most prevalent. Each year, approximately 400,000 children develop cancer. Treatment between countries vary, but usually includes either surgery, radiotherapy, or chemotherapy. Modern treatments such as hormone-, immuno- and antibody-based therapies are becoming increasingly popular. Several recent reports have been published on toxins, antibiotics, bacteriocins, non-ribosomal peptides, polyketides, phenylpropanoids, phenylflavonoids, purine nucleosides, short chain fatty acids (SCFAs) and enzymes with anticancer properties. Most of these molecules target cancer cells in a selective manner, either directly or indirectly through specific pathways. This review discusses the role of bacteria, including lactic acid bacteria, and their metabolites in the treatment of cancer.
Collapse
|
28
|
Mesothelin: An Immunotherapeutic Target beyond Solid Tumors. Cancers (Basel) 2022; 14:cancers14061550. [PMID: 35326701 PMCID: PMC8946840 DOI: 10.3390/cancers14061550] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/12/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This review summarizes the current knowledge on mesothelin’s function, its role in cancer, and opportunities for immunotherapeutic targeting of mesothelin. Immunotherapies including monoclonal antibodies, antibody–drug conjugates, chimeric antigen receptor T and NK-cells, targeted alpha therapies, and bispecific T cell engaging molecules are reviewed. We show future directions for mesothelin targeting in hematological malignancies, including acute myeloid leukemia. Abstract Modern targeted cancer therapies rely on the overexpression of tumor associated antigens with very little to no expression in normal cell types. Mesothelin is a glycosylphosphatidylinositol-anchored cell surface protein that has been identified in many different tumor types, including lung adenocarcinomas, ovarian carcinomas, and most recently in hematological malignancies, including acute myeloid leukemia (AML). Although the function of mesothelin is widely unknown, interactions with MUC16/CA125 indicate that mesothelin plays a role in the regulation of proliferation, growth, and adhesion signaling. Most research on mesothelin currently focuses on utilizing mesothelin to design targeted cancer therapies such as monoclonal antibodies, antibody–drug conjugates, chimeric antigen receptor T and NK cells, bispecific T cell engaging molecules, and targeted alpha therapies, amongst others. Both in vitro and in vivo studies using different immunotherapeutic modalities in mesothelin-positive AML models highlight the potential impact of this approach as a unique opportunity to treat hard-to-cure AML.
Collapse
|
29
|
Fortarezza F, Pezzuto F, Marzullo A, Cavone D, Romano DE, d'Amati A, Serio G, Vimercati L. Molecular Pathways in Peritoneal Mesothelioma: A Minireview of New Insights. Front Oncol 2022; 12:823839. [PMID: 35223506 PMCID: PMC8866824 DOI: 10.3389/fonc.2022.823839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
Mesothelioma is a rare malignant neoplasm with poor survival. It mainly affects the pleura (90%) but can arise in all serous cavities: peritoneum (5-10%), pericardium and tunica vaginalis testis (<1%). The onset of pleural mesothelioma is strictly related to asbestos exposure with a long latency time. The causal link with asbestos has also been suggested for peritoneal mesothelioma, while the importance of exposure in the onset of pericardial and tunica vaginalis testis mesotheliomas is not well known. Mesothelioma remains an aggressive and fatal disease with a five-year mortality rate higher than 95%. However, new therapeutic approaches based on molecular-targeted and immunomodulatory therapies are being explored but have conflicting results. In this context, the identification of critical targets appears mandatory. Awareness of the molecular and physiological changes leading to the neoplastic degeneration of mesothelial cells and the identification of gene mutations, epigenetic alterations, gene expression profiles and altered pathways could be helpful for selecting targetable mechanisms and molecules. In this review, we aimed to report recent research in the last 20 years focusing on the molecular pathways and prognostic factors in peritoneal mesothelioma and their possible diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Pathology Unit, University of Padova, Padova, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Pathology Unit, University of Padova, Padova, Italy
| | - Andrea Marzullo
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Domenica Cavone
- Department of Interdisciplinary Medicine, Occupational Health Unit, University of Bari, Bari, Italy
| | - Daniele Egidio Romano
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Antonio d'Amati
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Gabriella Serio
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Luigi Vimercati
- Department of Interdisciplinary Medicine, Occupational Health Unit, University of Bari, Bari, Italy
| |
Collapse
|
30
|
Bordeau BM, Abuqayyas L, Nguyen TD, Chen P, Balthasar JP. Development and Evaluation of Competitive Inhibitors of Trastuzumab-HER2 Binding to Bypass the Binding-Site Barrier. Front Pharmacol 2022; 13:837744. [PMID: 35250584 PMCID: PMC8895951 DOI: 10.3389/fphar.2022.837744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 12/31/2022] Open
Abstract
Our group has developed and experimentally validated a strategy to increase antibody penetration in solid tumors through transient inhibition of antibody-antigen binding. In prior work, we demonstrated that 1HE, an anti-trastuzumab single domain antibody that transiently inhibits trastuzumab binding to HER2, increased the penetration of trastuzumab and increased the efficacy of ado-trastuzumab emtansine (T-DM1) in HER2+ xenograft bearing mice. In the present work, 1HE variants were developed using random mutagenesis and phage display to enable optimization of tumor penetration and efficacy of trastuzumab-based therapeutics. To guide the rational selection of a particular 1HE mutant for a specific trastuzumab-therapy, we developed a mechanistic pharmacokinetic (PK) model to predict within-tumor exposure of trastuzumab/T-DM1. A pharmacodynamic (PD) component was added to the model to predict the relationship between intratumor exposure to T-DM1 and the corresponding therapeutic effect in HER2+ xenografts. To demonstrate the utility of the competitive inhibition approach for immunotoxins, PK parameters specific for a recombinant immunotoxin were incorporated into the model structure. Dissociation half-lives for variants ranged from 1.1 h (for variant LG11) to 107.9 h (for variant HE10). Simulations predicted that 1HE co-administration can increase the tumor penetration of T-DM1, with inhibitors with longer trastuzumab binding half-lives relative to 1HE (15.5 h) further increasing T-DM1 penetration at the expense of total tumor uptake of T-DM1. The PK/PD model accurately predicted the response of NCI-N87 xenografts to treatment with T-DM1 or T-DM1 co-administered with 1HE. Model predictions indicate that the 1HE mutant HF9, with a trastuzumab binding half-life of 51.1 h, would be the optimal inhibitor for increasing T-DM1 efficacy with a modest extension in the median survival time relative to T-DM1 with 1HE. Model simulations predict that LG11 co-administration will dramatically increase immunotoxin penetration within all tumor regions. We expect that the mechanistic model structure and the wide range of inhibitors developed in this work will enable optimization of trastuzumab-cytotoxin penetration and efficacy in solid tumors.
Collapse
Affiliation(s)
| | | | | | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
31
|
Shah R, Klotz LV, Glade J. Current Management and Future Perspective in Pleural Mesothelioma. Cancers (Basel) 2022; 14:1044. [PMID: 35205798 PMCID: PMC8869935 DOI: 10.3390/cancers14041044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/16/2022] Open
Abstract
Pleural mesothelioma is an aggressive malignancy arising from pleural mesothelial cell lining, predominantly associated with prior exposure to asbestos. The ban on asbestos use has led to its lower incidence in many countries, but globally the disease burden is expected to rise. Therefore, well-planned research is needed to develop more effective, tolerable and affordable drugs. The development of novel treatment has been too slow, with only two regimens of systemic therapy with robust phase 3 data approved formally to date. The treatment scenario for resectable disease remains controversial. However, recent developments in the understanding of disease and clinical trials have been encouraging, and may add better treatment options in the coming years. In this review, we discuss the current treatment options for pleural mesothelioma and shed light on some recent studies and ongoing trials.
Collapse
Affiliation(s)
- Rajiv Shah
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Laura V. Klotz
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University Hospital, 69126 Heidelberg, Germany;
| | - Julia Glade
- Institute for Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| |
Collapse
|
32
|
Havaei SM, Aucoin MG, Jahanian-Najafabadi A. Pseudomonas Exotoxin-Based Immunotoxins: Over Three Decades of Efforts on Targeting Cancer Cells With the Toxin. Front Oncol 2021; 11:781800. [PMID: 34976821 PMCID: PMC8716853 DOI: 10.3389/fonc.2021.781800] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the prominent causes of death worldwide. Despite the existence of various modalities for cancer treatment, many types of cancer remain uncured or develop resistance to therapeutic strategies. Furthermore, almost all chemotherapeutics cause a range of side effects because they affect normal cells in addition to malignant cells. Therefore, the development of novel therapeutic agents that are targeted specifically toward cancer cells is indispensable. Immunotoxins (ITs) are a class of tumor cell-targeted fusion proteins consisting of both a targeting moiety and a toxic moiety. The targeting moiety is usually an antibody/antibody fragment or a ligand of the immune system that can bind an antigen or receptor that is only expressed or overexpressed by cancer cells but not normal cells. The toxic moiety is usually a protein toxin (or derivative) of animal, plant, insect, or bacterial origin. To date, three ITs have gained Food and Drug Administration (FDA) approval for human use, including denileukin diftitox (FDA approval: 1999), tagraxofusp (FDA approval: 2018), and moxetumomab pasudotox (FDA approval: 2018). All of these ITs take advantage of bacterial protein toxins. The toxic moiety of the first two ITs is a truncated form of diphtheria toxin, and the third is a derivative of Pseudomonas exotoxin (PE). There is a growing list of ITs using PE, or its derivatives, being evaluated preclinically or clinically. Here, we will review these ITs to highlight the advances in PE-based anticancer strategies, as well as review the targeting moieties that are used to reduce the non-specific destruction of non-cancerous cells. Although we tried to be as comprehensive as possible, we have limited our review to those ITs that have proceeded to clinical trials and are still under active clinical evaluation.
Collapse
Affiliation(s)
- Seyed Mehdi Havaei
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marc G. Aucoin
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
33
|
Humeau J, Le Naour J, Galluzzi L, Kroemer G, Pol JG. Trial watch: intratumoral immunotherapy. Oncoimmunology 2021; 10:1984677. [PMID: 34676147 PMCID: PMC8526014 DOI: 10.1080/2162402x.2021.1984677] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
While chemotherapy and radiotherapy remain the first-line approaches for the management of most unresectable tumors, immunotherapy has emerged in the past two decades as a game-changing treatment, notably with the clinical success of immune checkpoint inhibitors. Immunotherapies aim at (re)activating anticancer immune responses which occur in two main steps: (1) the activation and expansion of tumor-specific T cells following cross-presentation of tumor antigens by specialized myeloid cells (priming phase); and (2) the immunological clearance of malignant cells by these antitumor T lymphocytes (effector phase). Therapeutic vaccines, adjuvants, monoclonal antibodies, cytokines, immunogenic cell death-inducing agents including oncolytic viruses, anthracycline-based chemotherapy and radiotherapy, as well as adoptive cell transfer, all act at different levels of this cascade to (re)instate cancer immunosurveillance. Intratumoral delivery of these immunotherapeutics is being tested in clinical trials to promote superior antitumor immune activity in the context of limited systemic toxicity.
Collapse
Affiliation(s)
- Juliette Humeau
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Institut Universitaire de France, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Jonathan G. Pol
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| |
Collapse
|
34
|
Khirehgesh MR, Sharifi J, Safari F, Akbari B. Immunotoxins and nanobody-based immunotoxins: review and update. J Drug Target 2021; 29:848-862. [PMID: 33615933 DOI: 10.1080/1061186x.2021.1894435] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunotoxins (ITs) are protein-based drugs that compose of targeting and cytotoxic moieties. After binding the IT to the specific cell-surface antigen, the IT internalises into the target cell and kills it. Targeting and cytotoxic moieties usually include monoclonal antibodies and protein toxins with bacterial or plant origin, respectively. ITs have been successful in haematologic malignancies treatment. However, ITs penetrate poorly into solid tumours because of their large size. Use of camelid antibody fragments known as nanobodies (Nbs) as a targeting moiety may overcome this problem. Nbs are the smallest fragment of antibodies with excellent tumour tissue penetration. The ability to recognise cryptic (immuno-evasive) target antigens, low immunogenicity, and high-affinity are other fundamental characteristics of Nbs that make them suitable candidates in targeted therapy. Here, we reviewed and discussed the structure and function of ITs, Nbs, and nanobody-based ITs. To gain sound insight into the issue at hand, we focussed on nanobody-based ITs.
Collapse
Affiliation(s)
- Mohammad Reza Khirehgesh
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jafar Sharifi
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Akbari
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
35
|
Dudnik E, Reinhorn D, Holtzman L. Novel and Promising Systemic Treatment Approaches in Mesothelioma. Curr Treat Options Oncol 2021; 22:89. [PMID: 34424409 DOI: 10.1007/s11864-021-00883-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2021] [Indexed: 10/20/2022]
Abstract
OPINION STATEMENT There was limited progress in the development of novel systemic approaches in the treatment of advanced malignant mesothelioma for years following the publication of the pivotal phase III trial of Vogelzang et al. that established the cisplatin/pemetrexed regimen as a standard 1st-line systemic therapy. Since then, over the last several years, a significant step forward has been made, with incorporation of immune checkpoint inhibitors and anti-angiogenic agents. In addition, better appreciation of mesothelioma biology has allowed detection of novelmolecular therapeutic targets. All the above-mentioned strategies, along with the additional promising approaches represented by adoptive T cell therapy, dendritic cell therapy, cancer vaccines, oncoviral therapy, and agents targeting mesothelin are discussed in this review. The clinical research to identify effective biologic targets and treatment combinations in malignant mesothelioma is ongoing.
Collapse
Affiliation(s)
- Elizabeth Dudnik
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, 49100, Petah Tikva, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, POB 39040 Ramat Aviv, 69978, Tel Aviv, Israel.
| | - Daniel Reinhorn
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, 49100, Petah Tikva, Israel
| | - Liran Holtzman
- Sackler Faculty of Medicine, Tel Aviv University, POB 39040 Ramat Aviv, 69978, Tel Aviv, Israel
| |
Collapse
|
36
|
Cui W, Popat S. Pleural mesothelioma (PM) - The status of systemic therapy. Cancer Treat Rev 2021; 100:102265. [PMID: 34399145 DOI: 10.1016/j.ctrv.2021.102265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/28/2021] [Accepted: 07/31/2021] [Indexed: 10/20/2022]
Abstract
Pleural mesothelioma (PM) remains a malignancy with poor prognosis. Despite initial disappointing response rates to single-agent chemotherapy, upfront platinum and anti-folate-based combination chemotherapy has remained the backbone of treatment for PM for the last three decades. The role of maintenance chemotherapy remains unclear; switch-maintenance gemcitabine has shown improvements in progression-free but not overall survival. The addition of antiangiogenic agents to chemotherapy yielded modest improvements in survival, both upfront in combination with platinum-pemetrexed, and in the relapsed setting. Immunotherapy, particularly PD-(L)1 inhibitors, has shown important but variable effectiveness in relapsed PM when used as monotherapy, and is an important salvage treatment after first-line chemotherapy. Furthermore, the randomized phase 3 trial of ipilimumab-nivolumab versus platinum-pemetrexed chemotherapy demonstrated improved overall survival favouring ipilimumab-nivolumab (HR 0.74, 96.6% CI 0.60-0.91; p = 0.0020), establishing this regimen as the new standard first-line treatment for PM, particularly in those with non-epithelioid histology. Increased interest in PM genomics has led to development of novel personalized therapeutics, such as those targeting DNA repair and EZH2 pathways, however with variable outcomes in trials. Targeting the membrane glycoprotein mesothelin and arginine deprivation are other important strategies under ongoing investigation. The field of PM is changing and new treatments bring hope to a largely lethal and poor prognostic malignancy. Despite these developments, current challenges include understanding the role of combination and multimodality treatments, drivers of resistance to treatment, and establishing predictive biomarkers to improve patient selection and treatment sequencing.
Collapse
Affiliation(s)
- Wanyuan Cui
- Lung Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Sanjay Popat
- Lung Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom; Thoracic Oncology, Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
37
|
Li Q, Li W, Xu K, Xing Y, Ding Y, Jing Z, Wang X, Hong Z. Preclinical evaluation of a novel anti-mesothelin immunotoxin based on a single domain antibody as the targeting ligand. Int J Pharm 2021; 602:120647. [PMID: 33915185 DOI: 10.1016/j.ijpharm.2021.120647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/21/2021] [Accepted: 04/22/2021] [Indexed: 01/10/2023]
Abstract
Pancreatic cancer, as one of the most aggressive and lethal malignancies in the world, is lack of effective treatment. Constructing immunotoxin molecules to target the mesothelin (MSLN) receptor is a potential therapeutic strategy for pancreatic cancer and other related malignant tumors, with some molecules being tested in clinical trials. However, currently, there are still some limitations in its applications, such as the difficulty of the preparation of drug molecules, the limited effectiveness of drugs, and the inadequacy of drug safety and immunogenicity. In this study, we constructed a novel type of anti-MSLN immunotoxin, A1-PE24X7, in which a single domain antibody (sdAb) molecule was used as the target ligand and an improved PE24X7 toxin with reduced off-target toxicity and immunogenicity was used as the effector. Unlike conventional immunotoxins, the designed A1-PE24X7 could be easily expressed in the E. coli system in the form of a soluble protein with a good yield (15--20 mg/L), avoiding the complex process of denaturation and refolding of inclusion bodies, and it can be conveniently stored in PBS solution for more than 7 days at 4 °C, showing high storage stability. Cell-based experiments showed that A1-PE24X7 entered MSLN-expressing tumor cells in a receptor-mediated manner and killed these cells with an EC50 in the low nanomolar range (0.13 nM against NCI-N87 cells and 0.79 nM against AsPC-1 cells) and it showed ideal selectivity for the MSLN receptor (>100 nM against receptor negative PC3 cells). In animal-based experiments, A1-PE24X7 had tumor enrichment ability in relation to MSLN-positive tumors and showed strong tumor killing and inhibition in mouse models of pancreatic cancer and gastric cancer. Five injections of 3.0 mg/kg A1-PE24X7 significantly reduced the tumor volume of gastric NCI-N87 cancer and also significantly inhibited the growth of pancreatic AsPC-1 cancer. In addition, the maximum tolerable dosage (MSD) of A1-PE24X7 to mice was higher than 15 mg/kg, showing that A1-PE24X7 has a relatively broad therapeutic window. These preclinical results indicate that this strategy has good potential for application to the treatment of pancreatic cancer and other tumors with high MSLN expression.
Collapse
Affiliation(s)
- Qiyu Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Wenjing Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Keyuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yutong Xing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yu Ding
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Zhe Jing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Xi Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
38
|
Mesothelin Expression in Human Tumors: A Tissue Microarray Study on 12,679 Tumors. Biomedicines 2021; 9:biomedicines9040397. [PMID: 33917081 PMCID: PMC8067734 DOI: 10.3390/biomedicines9040397] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/30/2021] [Accepted: 04/04/2021] [Indexed: 12/13/2022] Open
Abstract
Mesothelin (MSLN) represents an attractive molecule for targeted cancer therapies. To identify tumors that might benefit from such therapies, tissue microarrays including 15,050 tumors from 122 different tumor types and 76 healthy organs were analyzed for MSLN expression by immunohistochemistry. Sixty-six (54%) tumor types showed at least occasional weak staining, including 50 (41%) tumor types with at least one strongly positive sample. Highest prevalence of MSLN positivity had ovarian carcinomas (serous 97%, clear cell 83%, endometrioid 77%, mucinous 71%, carcinosarcoma 65%), pancreatic adenocarcinoma (ductal 75%, ampullary 81%), endometrial carcinomas (clear cell 71%, serous 57%, carcinosarcoma 50%, endometrioid 45%), malignant mesothelioma (69%), and adenocarcinoma of the lung (55%). MSLN was rare in cancers of the breast (7% of 1138), kidney (7% of 807), thyroid gland (1% of 638), soft tissues (0.3% of 931), and prostate (0 of 481). High expression was linked to advanced pathological tumor (pT) stage (p < 0.0001) and metastasis (p < 0.0001) in 1619 colorectal adenocarcinomas, but unrelated to parameters of malignancy in 1072 breast-, 386 ovarian-, 174 lung-, 757 kidney-, 171 endometrial-, 373 gastric-, and 925 bladder carcinomas. In summary, numerous important cancer types with high-level MSLN expression might benefit from future anti-MSLN therapies, but MSLN’s prognostic relevance appears to be limited.
Collapse
|
39
|
Hu ZI, Ghafoor A, Sengupta M, Hassan R. Malignant mesothelioma: Advances in immune checkpoint inhibitor and mesothelin-targeted therapies. Cancer 2021; 127:1010-1020. [PMID: 33620732 PMCID: PMC8555868 DOI: 10.1002/cncr.33433] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022]
Abstract
Malignant mesothelioma is an aggressive cancer with a poor prognosis and limited treatment options. For many years, the only US Food and Drug Administration-approved first-line treatment for unresectable mesothelioma was pemetrexed plus cisplatin. However, the recent approval of nivolumab plus ipilimumab as frontline treatment for patients with pleural mesothelioma marks a significant milestone for the treatment of this disease. In this review, the authors describe recent advances in therapeutic strategies for the treatment of patients with advanced, unresectable mesothelioma, highlighting the emerging use of immunotherapy and mesothelin-targeted therapies for the management of malignant mesothelioma.
Collapse
Affiliation(s)
- Zishuo I Hu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Azam Ghafoor
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Manjistha Sengupta
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|