1
|
Bocuzzi V, Bridoux J, Pirotte M, Withofs N, Hustinx R, D'Huyvetter M, Caers J, Marcion G. CD38 as theranostic target in oncology. J Transl Med 2024; 22:998. [PMID: 39501292 PMCID: PMC11539646 DOI: 10.1186/s12967-024-05768-6] [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: 07/23/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
CD38 is a multifunctional transmembrane glycoprotein found in multiple tissues and overexpressed in many cancer cells, notably in hematological malignancies such as leukemia and multiple myeloma (MM). Therefore, targeting CD38 remains an attractive strategy for cancer treatment in hematological malignancies as well as in solid tumors. It plays a critical role in the progression of these diseases through its ADP-ribosyl cyclase and cADPR-hydrolase activities. Its importance has led to the development of various anti-CD38 monoclonal antibodies (mAbs), including daratumumab and isatuximab, approved for MM treatment. These mAbs exert their anti-tumor effects through Fc-dependent immune mechanisms and immunomodulation, enhancing T-cell and NK-cell-mediated responses. However, resistance mechanisms arise during the treatment with daratumumab, creating the necessity for new therapies. This review explains current knowledge about the role of CD38 as a target in oncology and aims to delineate the use of single domain antibodies (sdAbs) as innovative theranostic tools in nuclear medicine. For diagnostic purposes, PET radionuclides like 68 Ga, 64Cu, and SPECT radionuclides like 99mTc and 111In, are commonly used. Significant progress has been made in anti-CD38 radioligand therapy (RLT), with anti-CD38 antibodies providing insights into tumor biology and treatment efficacy. In terms of therapy, RLT is a promising approach that offers precise targeting of malignant cells while minimizing exposure to healthy tissue. This involves the use of radionuclides emitting α particles, like 225Ac, 212Pb or 211At, and β--particles like 90Y, 131I, or 177Lu, to exert cytotoxic effects. Derived from Camelidae heavy chain antibodies, sdAbs offer advantages over conventional mAbs such as small size, high stability, specificity, and ability to recognize hidden epitopes. CD38-specific sdAbs, such as sdAb 2F8, characterized by our laboratory, showing excellent tumor targeting and their engineered constructs, such as biparatopic antibodies and chimeric antibodies, represent a new generation of theranostic agents for diagnosis and treatment CD38-expressing malignancies.
Collapse
Affiliation(s)
- Valentina Bocuzzi
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
- Center for Protein Engineering, University of Liège, Liège, Belgium
| | - Jessica Bridoux
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Nadia Withofs
- Department of Nuclear Medicine and Oncology, CHU de Liège, Liège, Belgium
| | - Roland Hustinx
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - Matthias D'Huyvetter
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jo Caers
- Department of Hematology, CHU de Liège, Liège, Belgium.
| | - Guillaume Marcion
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
- Center for Protein Engineering, University of Liège, Liège, Belgium
| |
Collapse
|
2
|
Zheleznyak A, Tang R, Duncan K, Manion B, Liang K, Xu B, Vanover A, Ghai A, Prior J, Lees S, Achilefu S, Kelly K, Shokeen M. Development of New CD38 Targeted Peptides for Cancer Imaging. Mol Imaging Biol 2024; 26:738-752. [PMID: 38480650 PMCID: PMC11282151 DOI: 10.1007/s11307-024-01901-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 04/18/2024]
Abstract
PURPOSE Multiple myeloma (MM) affects over 35,000 patients each year in the US. There remains a need for versatile Positron Emission Tomography (PET) tracers for the detection, accurate staging, and monitoring of treatment response of MM that have optimal specificity and translational attributes. CD38 is uniformly overexpressed in MM and thus represents an ideal target to develop CD38-targeted small molecule PET radiopharmaceuticals to address these challenges. PROCEDURES Using phage display peptide libraries and pioneering algorithms, we identified novel CD38 specific peptides. Imaging bioconjugates were synthesized using solid phase peptide chemistry, and systematically analyzed in vitro and in vivo in relevant MM systems. RESULTS The CD38-targeted bioconjugates were radiolabeled with copper-64 (64Cu) with100% radiochemical purity and an average specific activity of 3.3 - 6.6 MBq/nmol. The analog NODAGA-PEG4-SL022-GGS (SL022: Thr-His-Tyr-Pro-Ile-Val-Ile) had a Kd of 7.55 ± 0.291 nM and was chosen as the lead candidate. 64Cu-NODAGA-PEG4-SL022-GGS demonstrated high binding affinity to CD38 expressing human myeloma MM.1S-CBR-GFP-WT cells, which was blocked by the non-radiolabeled version of the peptide analog and anti-CD38 clinical antibodies, daratumumab and isatuximab, by 58%, 73%, and 78%, respectively. The CD38 positive MM.1S-CBR-GFP-WT cells had > 68% enhanced cellular binding when compared to MM.1S-CBR-GFP-KO cells devoid of CD38. Furthermore, our new CD38-targeted radiopharmaceutical allowed visualization of tumors located in marrow rich bones, remaining there for up to 4 h. Clearance from non-target organs occurred within 60 min. Quantitative PET data from a murine disseminated tumor model showed significantly higher accumulation in the bones of tumor-bearing animals compared to tumor-naïve animals (SUVmax 2.06 ± 0.4 versus 1.24 ± 0.4, P = 0.02). Independently, tumor uptake of the target compound was significantly higher (P = 0.003) compared to the scrambled peptide, 64Cu-NODAGA-PEG4-SL041-GGS (SL041: Thr-Tyr-His-Ile-Pro-Ile-Val). The subcutaneous MM model demonstrated significantly higher accumulation in tumors compared to muscle at 1 and 4 h after tracer administration (SUVmax 0.8 ± 0.2 and 0.14 ± 0.04, P = 0.04 at 1 h; SUVmax 0.89 ± 0.01 and 0.09 ± 0.01, P = 0.0002 at 4 h). CONCLUSIONS The novel CD38-targeted, radiolabeled bioconjugates were specific and allowed visualization of MM, providing a starting point for the clinical translation of such tracers for the detection of MM.
Collapse
Affiliation(s)
- Alexander Zheleznyak
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rui Tang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kathleen Duncan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brad Manion
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kexian Liang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Baogang Xu
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alexander Vanover
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Anchal Ghai
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Julie Prior
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Stephen Lees
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Samuel Achilefu
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kimberly Kelly
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Monica Shokeen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Alvin J. Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Wu Y, Zhu M, Sun B, Chen Y, Huang Y, Gai J, Li G, Li Y, Wan Y, Ma L. A humanized trivalent Nectin-4-targeting nanobody drug conjugate displays potent antitumor activity in gastric cancer. J Nanobiotechnology 2024; 22:256. [PMID: 38755613 PMCID: PMC11097425 DOI: 10.1186/s12951-024-02521-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Gastric cancer represents a highly lethal malignancy with an elevated mortality rate among cancer patients, coupled with a suboptimal postoperative survival prognosis. Nectin-4, an overexpressed oncological target for various cancers, has been exploited to create antibody-drug conjugates (ADCs) to treat solid tumors. However, there is limited research on Nectin-4 ADCs specifically for gastric cancer, and conventional immunoglobulin G (IgG)-based ADCs frequently encounter binding site barriers. Based on the excellent tumor penetration capabilities inherent in nanobodies (Nbs), we developed Nectin-4-targeting Nb drug conjugates (NDCs) for the treatment of gastric cancer. RESULTS An immunized phage display library was established and employed for the selection of Nectin-4-specific Nbs using phage display technology. Subsequently, these Nbs were engineered into homodimers to enhance Nb affinity. To prolong in vivo half-life and reduce immunogenicity, we fused an Nb targeting human serum albumin (HSA), resulting in the development of trivalent humanized Nbs. Further, we site-specifically conjugated a monomethyl auristatin E (MMAE) at the C-terminus of the trivalent Nbs, creating Nectin-4 NDC (huNb26/Nb26-Nbh-MMAE) with a drug-to-antibody ratio (DAR) of 1. Nectin-4 NDC demonstrated excellent in vitro cell-binding activities and cytotoxic efficacy against cells with high Nectin-4 expression. Subsequent administration of Nectin-4 NDC to mice bearing NCI-N87 human gastric cancer xenografts demonstrated rapid tissue penetration and high tumor uptake through in vivo imaging. Moreover, Nectin-4 NDC exhibited noteworthy dose-dependent anti-tumor efficacy in in vivo studies. CONCLUSION We have engineered a Nectin-4 NDC with elevated affinity and effective tumor uptake, further establishing its potential as a therapeutic agent for gastric cancer.
Collapse
Affiliation(s)
- Yue Wu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Min Zhu
- Shanghai Novamab Biopharmaceuticals Co., Ltd., Shanghai, China
| | - Baihe Sun
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yongting Chen
- Graduate School of Xinxiang Medical University, Henan, China
| | - Yuping Huang
- Shanghai Novamab Biopharmaceuticals Co., Ltd., Shanghai, China
| | - Junwei Gai
- Shanghai Novamab Biopharmaceuticals Co., Ltd., Shanghai, China
| | - Guanghui Li
- Shanghai Novamab Biopharmaceuticals Co., Ltd., Shanghai, China
| | - Yanfei Li
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China.
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co., Ltd., Shanghai, China.
| | - Linlin Ma
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China.
| |
Collapse
|
4
|
[Chinese expert consensus on the application of flow cytometry for the detectin of circulating plasma cells in plasma cell disorders (2024)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:313-321. [PMID: 38951057 PMCID: PMC11167996 DOI: 10.3760/cma.j.cn121090-20240117-00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Indexed: 07/03/2024]
Abstract
Flow cytometry plays an important role in the diagnosis and treatment of plasma cell diseases, particularly in the detection of circulating plasma cells (CPCs) in the peripheral blood. A consensus about the normalized use of flow cytometry in detection of CPCs in peripheral blood in clinical practice has been achieved. This consensus is founded on evidence-based principles, which elucidates the timing and value of flow cytometry for the detection of CPCs in the monoclonal gammopathy of undetermined significance, smoldering myeloma, multiple myeloma, and plasma cell leukemia and standardizes flow cytometry in the detection of CPCs in plasma cell diseases.
Collapse
|
5
|
Sharma AK, Gupta K, Mishra A, Lofland G, Marsh I, Kumar D, Ghiaur G, Imus P, Rowe SP, Hobbs RF, Gocke CB, Nimmagadda S. CD38-Specific Gallium-68 Labeled Peptide Radiotracer Enables Pharmacodynamic Monitoring in Multiple Myeloma with PET. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308617. [PMID: 38421139 PMCID: PMC11040352 DOI: 10.1002/advs.202308617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/09/2024] [Indexed: 03/02/2024]
Abstract
The limited availability of molecularly targeted low-molecular-weight imaging agents for monitoring multiple myeloma (MM)-targeted therapies has been a significant challenge in the field. In response, a first-in-class peptide-based radiotracer, [68Ga]Ga-AJ206, is developed that can be seamlessly integrated into the standard clinical workflow and is specifically designed to noninvasively quantify CD38 levels and pharmacodynamics by positron emission tomography (PET). A bicyclic peptide, AJ206, is synthesized and exhibits high affinity to CD38 (KD: 19.1 ± 0.99 × 10-9 m) by surface plasmon resonance. Further, [68Ga]Ga-AJ206-PET shows high contrast within 60 min and suitable absorbed dose estimates for clinical use. Additionally, [68Ga]Ga-AJ206 detects CD38 expression in cell line-derived xenografts, patient-derived xenografts (PDXs), and disseminated disease models in a manner consistent with flow cytometry and immunohistochemistry findings. Moreover, [68Ga]Ga-AJ206-PET successfully quantifies CD38 pharmacodynamics in PDXs, revealing increased CD38 expression in the tumor following all-trans retinoic acid (ATRA) therapy. In conclusion, [68Ga]Ga-AJ206 exhibits the salient features required for clinical translation, providing CD38-specific high-contrast images in multiple models of MM. [68Ga]Ga-AJ206-PET could be useful for quantifying total CD38 levels and pharmacodynamics during therapy to evaluate approved and new therapies in MM and other diseases with CD38 involvement.
Collapse
Affiliation(s)
- Ajay Kumar Sharma
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kuldeep Gupta
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Akhilesh Mishra
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Chemical & Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Gabriela Lofland
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ian Marsh
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Dhiraj Kumar
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Gabriel Ghiaur
- The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Philip Imus
- The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christian B Gocke
- The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|