1
|
Devasahayam Arokia Balaya R, Sen P, Grant CW, Zenka R, Sappani M, Lakshmanan J, Athreya AP, Kandasamy RK, Pandey A, Byeon SK. An integrative multi-omics analysis reveals a multi-analyte signature of pancreatic ductal adenocarcinoma in serum. J Gastroenterol 2024:10.1007/s00535-024-02197-6. [PMID: 39666045 DOI: 10.1007/s00535-024-02197-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 12/01/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a formidable health challenge due to its detection at a late stage and a lack of reliable biomarkers for early detection. Although levels of carbohydrate antigen 19-9 are often used in conjunction with imaging-based tests to aid in the diagnosis of PDAC, there is still a need for more sensitive and specific biomarkers for early detection of PDAC. METHODS We obtained serum samples from 88 subjects (patients with PDAC (n = 58) and controls (n = 30)). We carried out a multi-omics analysis to measure cytokines and related proteins using proximity extension technology and lipidomics and metabolomics using tandem mass spectrometry. Statistical analysis was carried out to find molecular alterations in patients with PDAC and a machine learning model was used to derive a molecular signature of PDAC. RESULTS We quantified 1,462 circulatory proteins along with 873 lipids and 1,001 metabolites. A total of 505 proteins, 186 metabolites and 33 lipids including bone marrow stromal antigen 2 (BST2), keratin 18 (KRT18), and cholesteryl ester(20:5) were found to be significantly altered in patients. We identified different levels of sphingosine, sphinganine, urobilinogen and lactose indicating that glycosphingolipid and galactose metabolisms were significantly altered in patients compared to controls. In addition, elevated levels of diacylglycerols and decreased cholesteryl esters were observed in patients. Using a machine learning model, we identified a signature of 38 biomarkers for PDAC, composed of 21 proteins, 4 lipids, and 13 metabolites. CONCLUSIONS Overall, this study identified several proteins, metabolites and lipids involved in various pathways including cholesterol and lipid metabolism to be changing in patients. In addition, we discovered a multi-analyte signature that could be further tested for detection of PDAC.
Collapse
Affiliation(s)
| | - Partho Sen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Caroline W Grant
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Roman Zenka
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marimuthu Sappani
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, 632002, India
| | - Jeyaseelan Lakshmanan
- College of Medicine, Mohammad Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, UAE
| | - Arjun P Athreya
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
Huang H, Pan Y, Mai Q, Zhang C, Du Q, Liao Y, Qin S, Chen Y, Huang J, Li J, Liu T, Zou Q, Zhou Y, Yuan L, Wang W, Liang Y, Pan CY, Liu J, Yao S. Targeting CDCP1 boost CD8+ T cells-mediated cytotoxicity in cervical cancer via the JAK/STAT signaling pathway. J Immunother Cancer 2024; 12:e009416. [PMID: 39455095 PMCID: PMC11529519 DOI: 10.1136/jitc-2024-009416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cervical cancer remains a global health challenge. The identification of new immunotherapeutic targets may provide a promising platform for advancing cervical cancer treatment. OBJECTIVE This study aims to investigate the role of CUB domain-containing protein 1 (CDCP1) in cervical cancer progression and evaluate its potential as a therapeutic target. METHODS We performed comprehensive analyses using patient cohorts and preclinical models to examine the association between CDCP1 expression and cervical cancer prognosis. Then in immunodeficient and immunocompetent mouse models, we further investigated the impact of CDCP1 on the tumor immune microenvironment, focusing on its effects on tumor-infiltrating T cells, including cytotoxic T lymphocytes (CTLs) and regulatory T cells (Tregs). Mechanistic studies were performed to elucidate the pathways involved in CDCP1-mediated immune modulation, in particular its interaction with the T cell receptor CD6 and the activation of the JAK-STAT signaling pathway. RESULTS Our results show that CDCP1 overexpression is associated with poor prognosis and T cell infliction in cervical cancer. Specifically, it affects the activity of CTLs and Tregs. Mechanistically, CDCP1 binds to CD6 and inhibits the JAK-STAT pathway of T cells. The study further demonstrates that targeting CDCP1 with the inhibitor 8-prenylnaringenin (8PN) effectively suppresses tumor growth in vivo and enhances antitumor immunity. CONCLUSIONS CDCP1 plays a critical role in cervical cancer progression by modulating the tumor immune microenvironment. Targeting CDCP1 offers a promising therapeutic strategy to improve the outcome of patients with cervical cancer.
Collapse
Affiliation(s)
- Hua Huang
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Yuwen Pan
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Qiuwen Mai
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Chunyu Zhang
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Yuandong Liao
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Shuhang Qin
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Yili Chen
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Jiaming Huang
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Jie Li
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Tianyu Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Qiaojian Zou
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Yijia Zhou
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Li Yuan
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Yanchun Liang
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Chao Yun Pan
- Department of Biochemistry, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Junxiu Liu
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Caravaca J, Bobba KN, Du S, Peter R, Gullberg GT, Bidkar AP, Flavell RR, Seo Y. A Technique to Quantify Very Low Activities in Regions of Interest With a Collimatorless Detector. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2745-2757. [PMID: 38478457 PMCID: PMC11293990 DOI: 10.1109/tmi.2024.3377142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
We present a new method to measure sub-microcurie activities of photon-emitting radionuclides in organs and lesions of small animals in vivo. Our technique, named the collimator-less likelihood fit, combines a very high sensitivity collimatorless detector with a Monte Carlo-based likelihood fit in order to estimate the activities in previously segmented regions of interest along with their uncertainties. This is done directly from the photon projections in our collimatorless detector and from the region of interest segmentation provided by an x-ray computed tomography scan. We have extensively validated our approach with 225Ac experimentally in spherical phantoms and mouse phantoms, and also numerically with simulations of a realistic mouse anatomy. Our method yields statistically unbiased results with uncertainties smaller than 20% for activities as low as ~111Bq (3nCi) and for exposures under 30 minutes. We demonstrate that our method yields more robust recovery coefficients when compared to SPECT imaging with a commercial pre-clinical scanner, specially at very low activities. Thus, our technique is complementary to traditional SPECT/CT imaging since it provides a more accurate and precise organ and tumor dosimetry, with a more limited spatial information. Finally, our technique is specially significant in extremely low-activity scenarios when SPECT/CT imaging is simply not viable.
Collapse
|
4
|
Shi Q, He J, Chen G, Xu J, Zeng Z, Zhao X, Zhao B, Gao X, Ye Z, Xiao M, Li H. The chemical composition of Diwu YangGan capsule and its potential inhibitory roles on hepatocellular carcinoma by microarray-based transcriptomics. J Tradit Complement Med 2024; 14:381-390. [PMID: 39035694 PMCID: PMC11259662 DOI: 10.1016/j.jtcme.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/28/2023] [Accepted: 12/24/2023] [Indexed: 07/23/2024] Open
Abstract
The Traditional Chinese Medicine compound preparation known as Diwu Yanggan capsule (DWYG) can effectively hinder the onset and progression of hepatocellular carcinoma (HCC), which is recognized worldwide as a significant contributor to fatalities associated with cancer. Nevertheless, the precise mechanisms implicated have remained ambiguous. In present study, the model of HCC was set up by the 2-acetylaminofluorene (2-AAF)/partial hepatectomy (PH) in rats. To confirm the differentially expressed genes (DEGs) identified in the microarray analysis, real-time quantitative reverse transcription PCR (qRT-PCR) was conducted. In the meantime, the liquid chromatography-quadrupole time of flight mass spectrometry (LC-QTOF-MS/MS) was employed to characterize the component profile of DWYG. Consequently, the DWYG treatment exhibited the ability to reverse 51 variation genes induced by 2-AAF/PH. Additionally, there was an overlap of 54 variation genes between the normal and model groups. Upon conducting RT-qPCR analysis, it was observed that the expression levels of all genes were increased by 2-AAF/PH and subsequently reversed after DWYG treatment. Notably, the fold change of expression levels for all genes was below 0.5, with 3 genes falling below 0.25. Moreover, an investigation was conducted to determine the signaling pathway that was activated/inhibited in the HCC group and subsequently reversed in the DWYG group. Moreover, the component profile of DWYG encompassed a comprehensive compilation of 206 compounds that were identified or characterized. The findings of this study elucidated the potential alleviative mechanisms of DWYG in the context of HCC, thereby holding significant implications for its future clinical utilization and widespread adoption.
Collapse
Affiliation(s)
- Qingxin Shi
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Jiangcheng He
- Wuhan Integrated Traditional Chinese and Western Medicine Orthopedic Hospital, Affiliated Hospital of Wuhan Sports University, Wuhan, 430079, China
| | - Guangya Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Jinlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zhaoxiang Zeng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xueyan Zhao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Binbin Zhao
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiang Gao
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Zhihua Ye
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Mingzhong Xiao
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Hanmin Li
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| |
Collapse
|
5
|
Lall R, Lee K, Chopra S, Kandala A, Evans M, Seo Y, Niknejad A, Anwar M. Low cost, high temporal resolution optical fiber-based γ-photon sensor for real-time pre-clinical evaluation of cancer-targeting radiopharmaceuticals. Biosens Bioelectron 2024; 247:115956. [PMID: 38145595 DOI: 10.1016/j.bios.2023.115956] [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: 10/06/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Cancer radiopharmaceutical therapies (RPTs) have demonstrated great promise in the treatment of neuroendocrine and prostate cancer, giving hope to late-stage metastatic cancer patients with currently very few treatment options. These therapies have sparked a large amount of interest in pre-clinical research due to their ability to target metastatic disease, with many research efforts focused towards developing and evaluating targeted RPTs for different cancer types in in vivo models. Here we describe a method for monitoring real-time in vivo binding kinetics for the pre-clinical evaluation of cancer RPTs. Recognizing the significant heterogeneity in biodistribution of RPTs among even genetically identical animal models, this approach offers long-term monitoring of the same in vivo organism without euthanasia in contrast to ex vivo tissue dosimetry, while providing high temporal resolution with a low-cost, easily assembled platform, that is not present in small-animal SPECT/CTs. The method utilizes the developed optical fiber-based γ-photon biosensor, characterized to have a wide linear dynamic range with Lutetium-177 (177Lu) activity (0.5-500 μCi/mL), a common radioisotope used in cancer RPT. The probe's ability to track in vivo uptake relative to SPECT/CT and ex vivo dosimetry techniques was verified by administering 177Lu-PSMA-617 to mouse models bearing human prostate cancer tumors (PC3-PIP, PC3-flu). With this method for monitoring RPT uptake, it is possible to evaluate changes in tissue uptake at temporal resolutions <1 min to determine RPT biodistribution in pre-clinical models and better understand dose relationships with tumor ablation, toxicity, and recurrence when attempting to move therapies towards clinical trial validation.
Collapse
Affiliation(s)
- Rahul Lall
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, 94720, USA.
| | - Kyoungtae Lee
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, 94107, USA
| | - Shalini Chopra
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94107, USA
| | - Averal Kandala
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Michael Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94107, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94107, USA
| | - Ali Niknejad
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Mekhail Anwar
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, 94720, USA; Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, 94107, USA
| |
Collapse
|
6
|
Ye X. Quantitative Membrane Proteomics for Discovery of Actionable Drug Targets at the Surface of RAS-Driven Human Cancer Cells. Methods Mol Biol 2024; 2823:27-46. [PMID: 39052212 DOI: 10.1007/978-1-0716-3922-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
With the advent of promising lung cancer immunotherapies targeting proteins at the cell surface of RAS-driven human cancers, the mass spectrometry (MS)-based surfaceomics remains a feasible strategy for therapeutic target discovery. This chapter describes a protocol for discovery of druggable protein targets at the surface of RAS-driven human cancer cells. This method relies on bottom-up MS-based quantitative surfaceomics that employs in parallel, targeted hydrazide-based cell-surface glycoproteomics and global shotgun membrane proteomics to enable unbiased quantitative profiling of thousands of cell surface membrane proteins. A large-scale molecular map of the KRASG12V surface was attained, resulting in confident detection and quantitation of more than 500 cell surface membrane proteins that were found to be unique or upregulated at the surface of cells harboring the KRASG12V mutant. A multistep bioinformatic progression revealed a subset of unique and/or significantly upregulated proteins as priority drug targets selected for orthogonal cross-validation using immunofluorescence, structured illumination microscopy, and western blotting. Among cross-validated targets, CUB domain containing protein 1 (CDCP1) and basigin (BSG-CD147) were selected as leading targets due to their involvement in cell adhesion and migration, consistent with the KRASG12V malignant phenotype as revealed by scanning electron microscopy and phenotypic cancer cell assays. Follow-up studies confirmed CDCP1 as an actionable therapeutic target, resulting in development of recombinant antibodies capable of killing KRAS-transformed cancer cells in preclinical setting. The present MS-based surfaceomics workflow represents a powerful drug target discovery platform that enables development of innovative immunotherapeutics (e.g., antibody drug conjugate against CDCP1) for attacking oncogenic RAS-driven cancers at the cell surface.
Collapse
Affiliation(s)
- Xiaoying Ye
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
7
|
Echavidre W, Fagret D, Faraggi M, Picco V, Montemagno C. Recent Pre-Clinical Advancements in Nuclear Medicine: Pioneering the Path to a Limitless Future. Cancers (Basel) 2023; 15:4839. [PMID: 37835533 PMCID: PMC10572076 DOI: 10.3390/cancers15194839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The theranostic approach in oncology holds significant importance in personalized medicine and stands as an exciting field of molecular medicine. Significant achievements have been made in this field in recent decades, particularly in treating neuroendocrine tumors using 177-Lu-radiolabeled somatostatin analogs and, more recently, in addressing prostate cancer through prostate-specific-membrane-antigen targeted radionuclide therapy. The promising clinical results obtained in these indications paved the way for the further development of this approach. With the continuous discovery of new molecular players in tumorigenesis, the development of novel radiopharmaceuticals, and the potential combination of theranostics agents with immunotherapy, nuclear medicine is poised for significant advancements. The strategy of theranostics in oncology can be categorized into (1) repurposing nuclear medicine agents for other indications, (2) improving existing radiopharmaceuticals, and (3) developing new theranostics agents for tumor-specific antigens. In this review, we provide an overview of theranostic development and shed light on its potential integration into combined treatment strategies.
Collapse
Affiliation(s)
- William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Daniel Fagret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, CHU Grenoble Alpes, Inserm, 38000 Grenoble, France;
| | - Marc Faraggi
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| |
Collapse
|
8
|
Frame E, Bobba K, Gunter D, Mihailescu L, Bidkar A, Flavell R, Vetter K. Coded aperture and Compton imaging for the development of 225 Ac-based radiopharmaceuticals. Med Phys 2023; 50:6454-6468. [PMID: 37672346 DOI: 10.1002/mp.16717] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/20/2023] [Accepted: 07/30/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Targeted alpha-particle therapy (TAT) has great promise as a cancer treatment. Arguably the most promising TAT radionuclide that has been proposed is 225 Ac. The development of 225 Ac-based radiopharmaceuticals has been hampered due to the lack of effective means to study the daughter redistribution of these agents in small animals at the preclinical stage. PURPOSE The ability to directly image the daughters, namely 221 Fr and 213 Bi, via their gamma-ray emissions would be a boon for preclinical studies. That said, conventional medical imaging modalities, including single photon emission computed tomography (SPECT) based on nonmultiplexed collimation, cannot be employed due to sensitivity limitations. METHODS As an alternative, we propose the use of both coded aperture and Compton imaging with the former modality suited to the 218-keV gamma-ray emission of 221 Fr and the latter suited to the 440-keV gamma-ray emission of 213 Bi. RESULTS This work includes coded aperture images of 221 Fr and Compton images of 213 Bi in tumor-bearing mice injected with 225 Ac-based radiopharmaceuticals. CONCLUSIONS These results are the first demonstration of visualizing and quantifying the 225 Ac daughters in small animals through the application of coded aperture and Compton imaging.
Collapse
Affiliation(s)
- Emily Frame
- Department of Nuclear Engineering, University of California Berkeley, Berkeley, California, USA
| | - Kondapa Bobba
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Donald Gunter
- Gunter Physics, Inc., Illinois, USA
- Applied Nuclear Physics Group, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | | | - Anil Bidkar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Robert Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Kai Vetter
- Department of Nuclear Engineering, University of California Berkeley, Berkeley, California, USA
- Applied Nuclear Physics Group, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| |
Collapse
|
9
|
Chopra S, Trepka K, Sakhamuri S, Carretero-González A, Zhu J, Egusa E, Zhou J, Leung K, Zhao N, Hooshdaran N, Feng FY, Wells JA, Chou J, Evans MJ. Theranostic Targeting of CUB Domain-Containing Protein 1 (CDCP1) in Multiple Subtypes of Bladder Cancer. Clin Cancer Res 2023; 29:1232-1242. [PMID: 36648492 PMCID: PMC10073270 DOI: 10.1158/1078-0432.ccr-22-1973] [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: 06/23/2022] [Revised: 11/13/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
PURPOSE Despite recent approvals for checkpoint inhibitors and antibody-drug conjugates targeting NECTIN4 or TROP2, metastatic bladder cancer remains incurable and new treatment strategies are urgently needed. CUB domain-containing protein 1 (CDCP1) is a cell surface protein and promising drug target for many cancers. This study aimed to determine whether CDCP1 is expressed in bladder cancer and whether CDCP1 can be targeted for treatment with radiolabeled antibodies. EXPERIMENTAL DESIGN CDCP1 expression was evaluated in four bladder cancer datasets (n = 1,047 biopsies). A tissue microarray of primary bladder cancer biopsies was probed for CDCP1 by IHC. CDCP1 expression was evaluated in patient-derived xenografts and cell lysates by immunoblot, flow cytometry, and saturation binding assays. Tumor detection in mouse bladder cancer models was tested using 89Zr-labeled 4A06, a monoclonal antibody targeting the ectodomain of CDCP1. 177Lu-4A06 was applied to mice bearing UMUC3 or HT-1376 xenografts to evaluate antitumor effects (CDCP1 expression in UMUC3 is 10-fold higher than HT-1376). RESULTS CDCP1 was highest in the basal/squamous subtype, and CDCP1 was expressed in 53% of primary biopsies. CDCP1 was not correlated with pathologic or tumor stage, metastatic site, or NECTIN4 and TROP2 at the mRNA or protein level. CDCP1 ranged from 105 to 106 receptors per cell. Mechanism studies showed that RAS signaling induced CDCP1 expression. 89Zr-4A06 PET detected five human bladder cancer xenografts. 177Lu-4A06 inhibited the growth of UMUC3 and HT-1376 xenografts, models with high and moderate CDCP1 expression, respectively. CONCLUSIONS These data establish that CDCP1 is expressed in bladder cancer, including TROP2 and NECTIN4-null disease, and suggest that bladder cancer can be treated with CDCP1-targeted radiotherapy.
Collapse
Affiliation(s)
- Shalini Chopra
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Kai Trepka
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
| | - Sasank Sakhamuri
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | | | - Jun Zhu
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
| | - Emily Egusa
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
| | - Jie Zhou
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Kevin Leung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Ning Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Nima Hooshdaran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Felix Y. Feng
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - James A. Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Jonathan Chou
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Michael J. Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
10
|
Kalita H, Patowary M. Biocompatible Polymer Nano-Constructs: A Potent Platform for Cancer Theranostics. Technol Cancer Res Treat 2023; 22:15330338231160391. [PMID: 36855787 PMCID: PMC9983094 DOI: 10.1177/15330338231160391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Nano-constructs of biocompatible polymers have drawn wide attention owing to their potential as theranostics for simultaneous therapy and detection of cancer. The present mini review summarizes various nano-architectures of polymers that have been developed as theranostic agents for the simultaneous treatment and diagnosis of cancer in a single platform. Additionally, research prospects of polymeric cancer theranostics for the future have been highlighted.
Collapse
Affiliation(s)
- Himani Kalita
- Department of Chemistry, 28678Indian Institute of Technology Guwahati, Guwahati, India.,Department of Chemistry, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Manoj Patowary
- School of Engineering, 560377MIT-ADT University, Pune, Maharashtra, India
| |
Collapse
|
11
|
Caravaca J, Huh Y, Gullberg GT, Seo Y. Compton and proximity imaging of 225Ac in vivo with a CZT gamma camera: a proof of principle with simulations. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2022; 6:904-915. [PMID: 36338821 PMCID: PMC9632644 DOI: 10.1109/trpms.2022.3166116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In vivo imaging of 225Ac is a major challenge in the development of targeted alpha therapy radiopharmaceuticals due to the extremely low injected doses. In this paper, we present the design of a multi-modality gamma camera that integrates both proximity and Compton imaging in order to achieve the demanding sensitivities required to image 225Ac with good image quality. We consider a dual-head camera, each of the heads consisting of two planar cadmium zinc telluride detectors acting as scatterer and absorber for Compton imaging, and with the scatterer practically in contact with the subject to allow for proximity imaging. We optimize the detector's design and characterize the detector's performance using Monte Carlo simulations. We show that Compton imaging can resolve features of up to 1.5 mm for hot rod phantoms with an activity of 1 μCi, and can reconstruct 3D images of a mouse injected with 0.5 μCi after a 15 minutes exposure and with a single bed position, for both 221Fr and 213Bi. Proximity imaging is able to resolve two 1 mm-radius sources of less than 0.1 μCi separated by 1 cm and at 1 mm from the detector, as well as it can provide planar images of 221Fr and 213Bi biodistributions of the mouse phantom in 5 minutes.
Collapse
Affiliation(s)
- Javier Caravaca
- Department of Radiology and Biomedical Imaging of the University of California San Francisco in San Francisco (CA) USA
| | - Yoonsuk Huh
- Department of Radiology and Biomedical Imaging of the University of California San Francisco in San Francisco (CA) USA
| | - Grant T Gullberg
- Department of Radiology and Biomedical Imaging of the University of California San Francisco in San Francisco (CA) USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging of the University of California San Francisco in San Francisco (CA) USA
| |
Collapse
|
12
|
Khan T, Lyons NJ, Gough M, Kwah KKX, Cuda TJ, Snell CE, Tse BW, Sokolowski KA, Pearce LA, Adams TE, Rose SE, Puttick S, Pajic M, Adams MN, He Y, Hooper JD, Kryza T. CUB Domain-Containing Protein 1 (CDCP1) is a rational target for the development of imaging tracers and antibody-drug conjugates for cancer detection and therapy. Am J Cancer Res 2022; 12:6915-6930. [PMID: 36276654 PMCID: PMC9576610 DOI: 10.7150/thno.78171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022] Open
Abstract
Rationale: An antibody-drug conjugate (ADC) is a targeted therapy consisting of a cytotoxic payload that is linked to an antibody which targets a protein enriched on malignant cells. Multiple ADCs are currently used clinically as anti-cancer agents significantly improving patient survival. Herein, we evaluated the rationale of targeting the cell surface oncoreceptor CUB domain-containing protein 1 (CDCP1) using ADCs and assessed the efficacy of CDCP1-directed ADCs against a range of malignant tumors. Methods: CDCP1 mRNA expression was evaluated using large transcriptomic datasets of normal/tumor samples for 23 types of cancer and 15 other normal organs, and CDCP1 protein expression was examined in 34 normal tissues, >300 samples from six types of cancer, and in 49 cancer cell lines. A recombinant human/mouse chimeric anti-CDCP1 antibody (ch10D7) was labelled with 89Zirconium or monomethyl auristatin E (MMAE) and tested in multiple pre-clinical cancer models including 36 cancer cell lines and three mouse xenograft models. Results: Analysis of CDCP1 expression indicates elevated CDCP1 expression in the majority of the cancers and restricted expression in normal human tissues. Antibody ch10D7 demonstrates a high affinity and specificity for CDCP1 inducing cell signalling via Src accompanied by rapid internalization of ch10D7/CDCP1 complexes in cancer cells.89Zirconium-labelled ch10D7 accumulates in CDCP1 expressing cells enabling detection of pancreatic cancer xenografts in mice by PET imaging. Cytotoxicity of MMAE-labelled ch10D7 against kidney, colorectal, lung, ovarian, pancreatic and prostate cancer cells in vitro, correlates with the level of CDCP1 on the plasma membrane. ch10D7-MMAE displays robust anti-tumor effects against mouse xenograft models of pancreatic, colorectal and ovarian cancer. Conclusion: CDCP1 directed imaging agents will be useful for selecting cancer patients for personalized treatment with cytotoxin-loaded CDCP1 targeting agents including antibody-drug conjugates.
Collapse
Affiliation(s)
- Tashbib Khan
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - Nicholas J Lyons
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - Madeline Gough
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - Kayden K X Kwah
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - Tahleesa J Cuda
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - Cameron E Snell
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia.,Mater Health Services, South Brisbane, QLD, Australia
| | - Brian W Tse
- Preclinical Imaging Facility, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Lesley A Pearce
- Commonwealth Scientific and Industrial Research Organisation Manufacturing, Parkville, VIC, Australia
| | - Timothy E Adams
- Commonwealth Scientific and Industrial Research Organisation Manufacturing, Parkville, VIC, Australia
| | - Stephen E Rose
- Commonwealth Scientific and Industrial Research Organisation, Herston, QLD, Australia
| | - Simon Puttick
- Commonwealth Scientific and Industrial Research Organisation, Herston, QLD, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Mark N Adams
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| | - Thomas Kryza
- Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, Australia
| |
Collapse
|
13
|
Zhao N, Chopra S, Trepka K, Wang YH, Sakhamuri S, Hooshdaran N, Kim H, Zhuo J, Lim SA, Leung KK, Egusa EA, Zhu J, Zhang L, Foye A, Sriram R, Chan E, Seo Y, Feng FY, Small EJ, Chou J, Wells JA, Aggarwal R, Evans MJ. CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease. Clin Cancer Res 2022; 28:3066-3075. [PMID: 35604681 PMCID: PMC9288514 DOI: 10.1158/1078-0432.ccr-21-3858] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/07/2022] [Accepted: 05/17/2022] [Indexed: 01/17/2023]
Abstract
PURPOSE With the improvement in overall survival with 177Lu-PSMA 617, radioligand therapy (RLT) is now a viable option for patients with metastatic castration-resistant prostate cancer (mCRPC). However, responses are variable, in part due to low PSMA expression in 30% of patients. Herein, we evaluated whether the cell surface protein CUB domain-containing protein 1 (CDCP1) can be exploited to treat mCRPC with RLT, including in PSMA-low subsets. EXPERIMENTAL DESIGN CDCP1 levels were evaluated using RNA sequencing from 119 mCRPC biopsies. CDCP1 levels were assessed in 17 post-enzalutamide- or abiraterone-treated mCRPC biopsies, 12 patient-derived xenografts (PDX), and prostate cancer cell lines. 4A06, a recombinant human antibody that targets the CDCP1 ectodomain, was labeled with Zr-89 or Lu-177 and tested in tumor-bearing mice. RESULTS CDCP1 expression was observed in 90% of mCRPC biopsies, including small-cell neuroendocrine (SCNC) and adenocarcinomas with low FOLH1 (PSMA) levels. Fifteen of 17 evaluable mCRPC biopsies (85%) demonstrated membranous CDCP1 expression, and 4 of 17 (23%) had higher CDCP1 H-scores compared with PSMA. CDCP1 was expressed in 10 of 12 PDX samples. Bmax values of approximately 22,000, 6,200, and 2,800 fmol/mg were calculated for PC3, DU145, and C4-2B human prostate cancer cells, respectively. 89Zr-4A06 PET detected six human prostate cancer xenografts, including PSMA-low tumors. 177Lu-4A06 significantly suppressed growth of DU145 and C4-2B xenografts. CONCLUSIONS The data provide the first evidence supporting CDCP1-directed RLT to treat mCRPC. Expanded studies are warranted to determine whether CDCP1 is a viable drug target for patients with mCPRC.
Collapse
Affiliation(s)
- Ning Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Shalini Chopra
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Kai Trepka
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158
| | - Yung-hua Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Sasank Sakhamuri
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Nima Hooshdaran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Hyunjung Kim
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Jie Zhuo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Shion A. Lim
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Kevin K. Leung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158
| | - Emily A. Egusa
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Jun Zhu
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Li Zhang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158
| | - Adam Foye
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Emily Chan
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158
| | - Felix Y. Feng
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Eric J. Small
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Jonathan Chou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - James A. Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Rahul Aggarwal
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| | - Michael J. Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
14
|
CDCP1: A promising diagnostic biomarker and therapeutic target for human cancer. Life Sci 2022; 301:120600. [DOI: 10.1016/j.lfs.2022.120600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
|
15
|
Muir RK, Guerra M, Bogyo MM. Activity-Based Diagnostics: Recent Advances in the Development of Probes for Use with Diverse Detection Modalities. ACS Chem Biol 2022; 17:281-291. [PMID: 35026106 DOI: 10.1021/acschembio.1c00753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abnormal enzyme expression and activity is a hallmark of many diseases. Activity-based diagnostics are a class of chemical probes that aim to leverage this dysregulated metabolic signature to produce a detectable signal specific to diseased tissue. In this Review, we highlight recent methodologies employed in activity-based diagnostics that provide exquisite signal sensitivity and specificity in complex biological systems for multiple disease states. We divide these examples based upon their unique signal readout modalities and highlight those that have advanced into clinical trials.
Collapse
Affiliation(s)
- Ryan K. Muir
- Department of Pathology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Matteo Guerra
- Department of Pathology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Matthew M. Bogyo
- Department of Pathology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
16
|
Lim SA, Zhou J, Martinko AJ, Wang YH, Filippova EV, Steri V, Wang D, Remesh SG, Liu J, Hann B, Kossiakoff AA, Evans MJ, Leung KK, Wells JA. Targeting a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) for RAS-driven cancers. J Clin Invest 2022; 132:e154604. [PMID: 35166238 PMCID: PMC8843743 DOI: 10.1172/jci154604] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective and safer treatments for solid tumors. CDCP1 is highly overexpressed in RAS-driven cancers, and its ectodomain is cleaved by extracellular proteases. Biochemical, biophysical, and structural characterization revealed that the 2 cleaved fragments of CDCP1 remain tightly associated with minimal proteolysis-induced conformational change. Using differential phage display, we generated recombinant Abs that are exquisitely selective to cleaved CDCP1 with no detectable binding to the uncleaved form. These Abs potently targeted cleaved CDCP1-expressing cancer cells as an Ab-drug conjugate, an Ab-radionuclide conjugate, and a bispecific T cell engager. In a syngeneic pancreatic tumor model, these cleaved-specific Abs showed tumor-specific localization and antitumor activity with superior safety profiles compared with a pan-CDCP1 approach. Targeting proteolytic neoepitopes could provide an orthogonal "AND" gate for improving the therapeutic index.
Collapse
Affiliation(s)
| | - Jie Zhou
- Department of Pharmaceutical Chemistry
| | | | - Yung-Hua Wang
- Department of Radiology and Biomedical Imaging, and
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Ekaterina V. Filippova
- Department of Biochemistry and Molecular Biology, and
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, USA
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Preclinical Therapeutics Core, UCSF, San Francisco, California, USA
| | - Donghui Wang
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Preclinical Therapeutics Core, UCSF, San Francisco, California, USA
| | | | - Jia Liu
- Department of Pharmaceutical Chemistry
| | - Byron Hann
- Preclinical Therapeutics Core, UCSF, San Francisco, California, USA
| | - Anthony A. Kossiakoff
- Department of Biochemistry and Molecular Biology, and
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, USA
| | - Michael J. Evans
- Department of Radiology and Biomedical Imaging, and
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | | | - James A. Wells
- Department of Pharmaceutical Chemistry
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, California, USA
| |
Collapse
|
17
|
Donahue KL, Pasca di Magliano M. Cleaved CDCP1 marks the spot: a neoepitope for RAS-driven cancers. J Clin Invest 2022; 132:e157168. [PMID: 35166242 PMCID: PMC8843638 DOI: 10.1172/jci157168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A challenge in cancer treatment is targeting cancer cells while sparing normal cells. Thus, identifying cancer-specific neoepitopes is an active research area. Neoepitopes are generated by the accumulation of mutations; however, deadly cancer types, including pancreatic cancer, have a low mutational burden and, consequently, a paucity of neoantigens. In this issue of the JCI, Lim, Zhou, and colleagues describe a neoepitope generated upon proteolytic cleavage of the transmembrane CUB domain containing protein 1 (CDCP1). CDCP1 is overexpressed in cancer and portends a worse prognosis; previous attempts to target CDCP1 reduced cancer growth, but adversely affected the host. Here, the authors generated an antibody that specifically targeted cleaved CDCP1 (c-CDCP1) and developed a drug conjugate, a vector for radioactive ions, and a mediator of T cell activation. The therapeutics inhibited pancreatic cancer cell growth in vitro and in vivo. Exploiting proteolytic cleavage-derived neoantigens opens an attractive way for specifically targeting cancer cells.
Collapse
Affiliation(s)
| | - Marina Pasca di Magliano
- Department of Surgery, Department of Cell and Developmental Biology, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
18
|
Kumar AA, Buckley BJ, Ranson M. The Urokinase Plasminogen Activation System in Pancreatic Cancer: Prospective Diagnostic and Therapeutic Targets. Biomolecules 2022; 12:152. [PMID: 35204653 PMCID: PMC8961517 DOI: 10.3390/biom12020152] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy that features high recurrence rates and the poorest prognosis of all solid cancers. The urokinase plasminogen activation system (uPAS) is strongly implicated in the pathophysiology and clinical outcomes of patients with pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic cancers. Overexpression of the urokinase-type plasminogen activator (uPA) or its cell surface receptor uPAR is a key step in the acquisition of a metastatic phenotype via multiple mechanisms, including the increased activation of cell surface localised plasminogen which generates the serine protease plasmin. This triggers multiple downstream processes that promote tumour cell migration and invasion. Increasing clinical evidence shows that the overexpression of uPA, uPAR, or of both is strongly associated with worse clinicopathological features and poor prognosis in PDAC patients. This review provides an overview of the current understanding of the uPAS in the pathogenesis and progression of pancreatic cancer, with a focus on PDAC, and summarises the substantial body of evidence that supports the role of uPAS components, including plasminogen receptors, in this disease. The review further outlines the clinical utility of uPAS components as prospective diagnostic and prognostic biomarkers for PDAC, as well as a rationale for the development of novel uPAS-targeted therapeutics.
Collapse
Affiliation(s)
- Ashna A. Kumar
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Benjamin J. Buckley
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
19
|
van Dam MA, Vuijk FA, Stibbe JA, Houvast RD, Luelmo SAC, Crobach S, Shahbazi Feshtali S, de Geus-Oei LF, Bonsing BA, Sier CFM, Kuppen PJK, Swijnenburg RJ, Windhorst AD, Burggraaf J, Vahrmeijer AL, Mieog JSD. Overview and Future Perspectives on Tumor-Targeted Positron Emission Tomography and Fluorescence Imaging of Pancreatic Cancer in the Era of Neoadjuvant Therapy. Cancers (Basel) 2021; 13:6088. [PMID: 34885196 PMCID: PMC8656821 DOI: 10.3390/cancers13236088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite recent advances in the multimodal treatment of pancreatic ductal adenocarcinoma (PDAC), overall survival remains poor with a 5-year cumulative survival of approximately 10%. Neoadjuvant (chemo- and/or radio-) therapy is increasingly incorporated in treatment strategies for patients with (borderline) resectable and locally advanced disease. Neoadjuvant therapy aims to improve radical resection rates by reducing tumor mass and (partial) encasement of important vascular structures, as well as eradicating occult micrometastases. Results from recent multicenter clinical trials evaluating this approach demonstrate prolonged survival and increased complete surgical resection rates (R0). Currently, tumor response to neoadjuvant therapy is monitored using computed tomography (CT) following the RECIST 1.1 criteria. Accurate assessment of neoadjuvant treatment response and tumor resectability is considered a major challenge, as current conventional imaging modalities provide limited accuracy and specificity for discrimination between necrosis, fibrosis, and remaining vital tumor tissue. As a consequence, resections with tumor-positive margins and subsequent early locoregional tumor recurrences are observed in a substantial number of patients following surgical resection with curative intent. Of these patients, up to 80% are diagnosed with recurrent disease after a median disease-free interval of merely 8 months. These numbers underline the urgent need to improve imaging modalities for more accurate assessment of therapy response and subsequent re-staging of disease, thereby aiming to optimize individual patient's treatment strategy. In cases of curative intent resection, additional intra-operative real-time guidance could aid surgeons during complex procedures and potentially reduce the rate of incomplete resections and early (locoregional) tumor recurrences. In recent years intraoperative imaging in cancer has made a shift towards tumor-specific molecular targeting. Several important molecular targets have been identified that show overexpression in PDAC, for example: CA19.9, CEA, EGFR, VEGFR/VEGF-A, uPA/uPAR, and various integrins. Tumor-targeted PET/CT combined with intraoperative fluorescence imaging, could provide valuable information for tumor detection and staging, therapy response evaluation with re-staging of disease and intraoperative guidance during surgical resection of PDAC. METHODS A literature search in the PubMed database and (inter)national trial registers was conducted, focusing on studies published over the last 15 years. Data and information of eligible articles regarding PET/CT as well as fluorescence imaging in PDAC were reviewed. Areas covered: This review covers the current strategies, obstacles, challenges, and developments in targeted tumor imaging, focusing on the feasibility and value of PET/CT and fluorescence imaging for integration in the work-up and treatment of PDAC. An overview is given of identified targets and their characteristics, as well as the available literature of conducted and ongoing clinical and preclinical trials evaluating PDAC-targeted nuclear and fluorescent tracers.
Collapse
Affiliation(s)
- Martijn A. van Dam
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Floris A. Vuijk
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Judith A. Stibbe
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Ruben D. Houvast
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Saskia A. C. Luelmo
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Stijn Crobach
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, University Medical Center Leiden, 2333 ZA Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | | | - Albert D. Windhorst
- Department of Radiology, Section of Nuclear Medicine, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, The Netherlands;
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Centre for Human Drug Research, 2333 CL Leiden, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| |
Collapse
|
20
|
Kaczmarczyk JA, Roberts RR, Luke BT, Chan KC, Van Wagoner CM, Felder RA, Saul RG, Simona C, Blonder J. Comparative microsomal proteomics of a model lung cancer cell line NCI-H23 reveals distinct differences between molecular profiles of 3D and 2D cultured cells. Oncotarget 2021; 12:2022-2038. [PMID: 34611477 PMCID: PMC8487723 DOI: 10.18632/oncotarget.28072] [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: 06/15/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the USA and worldwide. Yet, about 95% of new drug candidates validated in preclinical phase eventually fail in clinical trials. Such a high attrition rate is attributed mostly to the inability of conventional two-dimensionally (2D) cultured cancer cells to mimic native three-dimensional (3D) growth of malignant cells in human tumors. To ascertain phenotypical differences between these two distinct culture conditions, we carried out a comparative proteomic analysis of a membrane fraction obtained from 3D- and 2D-cultured NSCLC model cell line NCI-H23. This analysis revealed a map of 1,166 (24%) protein species regulated in culture dependent manner, including differential regulation of a subset of cell surface-based CD molecules. We confirmed exclusive expression of CD99, CD146 and CD239 in 3D culture. Furthermore, label-free quantitation, targeting KRas proteoform-specific peptides, revealed upregulation of both wild type and monoallelic KRas4BG12C mutant at the surface of 3D cultured cells. In order to reduce the high attrition rate of new drug candidates, the results of this study strongly suggests exploiting base-line molecular profiling of a large number of patient-derived NSCLC cell lines grown in 2D and 3D culture, prior to actual drug candidate testing.
Collapse
Affiliation(s)
- Jan A. Kaczmarczyk
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Rhonda R. Roberts
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Brian T. Luke
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - King C. Chan
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Current address: The Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Carly M. Van Wagoner
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Current address: The Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Robin A. Felder
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Richard G. Saul
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Colantonio Simona
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Josip Blonder
- Antibody Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| |
Collapse
|
21
|
Montemagno C, Cassim S, De Leiris N, Durivault J, Faraggi M, Pagès G. Pancreatic Ductal Adenocarcinoma: The Dawn of the Era of Nuclear Medicine? Int J Mol Sci 2021; 22:6413. [PMID: 34203923 PMCID: PMC8232627 DOI: 10.3390/ijms22126413] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), accounting for 90-95% of all pancreatic tumors, is a highly devastating disease associated with poor prognosis. The lack of accurate diagnostic tests and failure of conventional therapies contribute to this pejorative issue. Over the last decade, the advent of theranostics in nuclear medicine has opened great opportunities for the diagnosis and treatment of several solid tumors. Several radiotracers dedicated to PDAC imaging or internal vectorized radiotherapy have been developed and some of them are currently under clinical consideration. The functional information provided by Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) could indeed provide an additive diagnostic value and thus help in the selection of patients for targeted therapies. Moreover, the therapeutic potential of β-- and α-emitter-radiolabeled agents could also overcome the resistance to conventional therapies. This review summarizes the current knowledge concerning the recent developments in the nuclear medicine field for the management of PDAC patients.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and IN-SERM U1081, Université Cote d’Azur, 06200 Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Shamir Cassim
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Nicolas De Leiris
- Nuclear Medicine Department, Grenoble-Alpes University Hospital, 38000 Grenoble, France;
- Laboratoire Radiopharmaceutiques Biocliniques, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France
| | - Jérôme Durivault
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Marc Faraggi
- Centre Hospitalier Princesse Grace, Nuclear Medicine Department, 98000 Monaco, Monaco;
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and IN-SERM U1081, Université Cote d’Azur, 06200 Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| |
Collapse
|
22
|
Arias-Pinilla GA, Modjtahedi H. Therapeutic Application of Monoclonal Antibodies in Pancreatic Cancer: Advances, Challenges and Future Opportunities. Cancers (Basel) 2021; 13:1781. [PMID: 33917882 PMCID: PMC8068268 DOI: 10.3390/cancers13081781] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer remains as one of the most aggressive cancer types. In the absence of reliable biomarkers for its early detection and more effective therapeutic interventions, pancreatic cancer is projected to become the second leading cause of cancer death in the Western world in the next decade. Therefore, it is essential to discover novel therapeutic targets and to develop more effective and pancreatic cancer-specific therapeutic agents. To date, 45 monoclonal antibodies (mAbs) have been approved for the treatment of patients with a wide range of cancers; however, none has yet been approved for pancreatic cancer. In this comprehensive review, we discuss the FDA approved anticancer mAb-based drugs, the results of preclinical studies and clinical trials with mAbs in pancreatic cancer and the factors contributing to the poor response to antibody therapy (e.g. tumour heterogeneity, desmoplastic stroma). MAb technology is an excellent tool for studying the complex biology of pancreatic cancer, to discover novel therapeutic targets and to develop various forms of antibody-based therapeutic agents and companion diagnostic tests for the selection of patients who are more likely to benefit from such therapy. These should result in the approval and routine use of antibody-based agents for the treatment of pancreatic cancer patients in the future.
Collapse
Affiliation(s)
- Gustavo A. Arias-Pinilla
- Department of Oncology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK;
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| | - Helmout Modjtahedi
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| |
Collapse
|
23
|
van der Weyden L, Harle V, Turner G, Offord V, Iyer V, Droop A, Swiatkowska A, Rabbie R, Campbell AD, Sansom OJ, Pardo M, Choudhary JS, Ferreira I, Tullett M, Arends MJ, Speak AO, Adams DJ. CRISPR activation screen in mice identifies novel membrane proteins enhancing pulmonary metastatic colonisation. Commun Biol 2021; 4:395. [PMID: 33758365 PMCID: PMC7987976 DOI: 10.1038/s42003-021-01912-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/25/2021] [Indexed: 02/08/2023] Open
Abstract
Melanoma represents ~5% of all cutaneous malignancies, yet accounts for the majority of skin cancer deaths due to its propensity to metastasise. To develop new therapies, novel target molecules must to be identified and the accessibility of cell surface proteins makes them attractive targets. Using CRISPR activation technology, we screened a library of guide RNAs targeting membrane protein-encoding genes to identify cell surface molecules whose upregulation enhances the metastatic pulmonary colonisation capabilities of tumour cells in vivo. We show that upregulated expression of the cell surface protein LRRN4CL led to increased pulmonary metastases in mice. Critically, LRRN4CL expression was elevated in melanoma patient samples, with high expression levels correlating with decreased survival. Collectively, our findings uncover an unappreciated role for LRRN4CL in the outcome of melanoma patients and identifies a potential therapeutic target and biomarker.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- CRISPR-Cas Systems
- Cell Line, Tumor
- Cell Movement
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Male
- Melanoma, Experimental/genetics
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/secondary
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Neoplasm Invasiveness
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Up-Regulation
Collapse
Affiliation(s)
| | - Victoria Harle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Gemma Turner
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Victoria Offord
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Vivek Iyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Alastair Droop
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Roy Rabbie
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Ingrid Ferreira
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Mark Tullett
- Western Sussex NHS Foundation Trust, Chichester, West Sussex, UK
| | - Mark J Arends
- University of Edinburgh Division of Pathology, Edinburgh Cancer Research UK Cancer Centre, Institute of Genetics & Molecular Medicine, Edinburgh, UK
| | - Anneliese O Speak
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - David J Adams
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
24
|
Khan T, Kryza T, Lyons NJ, He Y, Hooper JD. The CDCP1 Signaling Hub: A Target for Cancer Detection and Therapeutic Intervention. Cancer Res 2021; 81:2259-2269. [PMID: 33509939 DOI: 10.1158/0008-5472.can-20-2978] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
CUB-domain containing protein 1 (CDCP1) is a type I transmembrane glycoprotein that is upregulated in malignancies of the breast, lung, colorectum, ovary, kidney, liver, pancreas, and hematopoietic system. Here, we discuss CDCP1 as an important hub for oncogenic signaling and its key roles in malignant transformation and summarize approaches focused on exploiting it for cancer diagnosis and therapy. Elevated levels of CDCP1 are associated with progressive disease and markedly poorer survival. Predominantly located on the cell surface, CDCP1 lies at the nexus of key tumorigenic and metastatic signaling cascades, including the SRC/PKCδ, PI3K/AKT, WNT, and RAS/ERK axes, the oxidative pentose phosphate pathway, and fatty acid oxidation, making important functional contributions to cancer cell survival and growth, metastasis, and treatment resistance. These findings have stimulated the development of agents that target CDCP1 for detection and treatment of a range of cancers, and results from preclinical models suggest that these approaches could be efficacious and have manageable toxicity profiles.
Collapse
Affiliation(s)
- Tashbib Khan
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas J Lyons
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
25
|
Birnbaum DJ, Finetti P, Birnbaum D, Bertucci F. Theranostic Targeting of CUB Domain Containing Protein 1 (CDCP1) in Pancreatic Cancer—Letter. Clin Cancer Res 2020; 26:5539. [DOI: 10.1158/1078-0432.ccr-20-1969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/02/2020] [Accepted: 08/07/2020] [Indexed: 11/16/2022]
Affiliation(s)
- David J. Birnbaum
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Daniel Birnbaum
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - François Bertucci
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| |
Collapse
|