1
|
Shraim R, Mooney B, Conkrite KL, Weiner AK, Morin GB, Sorensen PH, Maris JM, Diskin SJ, Sacan A. IMMUNOTAR - Integrative prioritization of cell surface targets for cancer immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597422. [PMID: 38895237 PMCID: PMC11185603 DOI: 10.1101/2024.06.04.597422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Cancer remains a leading cause of mortality globally. Recent improvements in survival have been facilitated by the development of less toxic immunotherapies; however, identifying targets for immunotherapies remains a challenge in the field. To address this challenge, we developed IMMUNOTAR, a computational tool that systematically prioritizes and identifies candidate immunotherapeutic targets. IMMUNOTAR integrates user-provided RNA-sequencing or proteomics data with quantitative features extracted from publicly available databases based on predefined optimal immunotherapeutic target criteria and quantitatively prioritizes potential surface protein targets. We demonstrate the utility and flexibility of IMMUNOTAR using three distinct datasets, validating its effectiveness in identifying both known and new potential immunotherapeutic targets within the analyzed cancer phenotypes. Overall, IMMUNOTAR enables the compilation of data from multiple sources into a unified platform, allowing users to simultaneously evaluate surface proteins across diverse criteria. By streamlining target identification, IMMUNOTAR empowers researchers to efficiently allocate resources and accelerate immunotherapy development.
Collapse
Affiliation(s)
- Rawan Shraim
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- School of Biomedical Engineering, Science and Health System, Drexel University, Philadelphia, PA 19104, USA
| | - Brian Mooney
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Karina L. Conkrite
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Amber K. Weiner
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Gregg B. Morin
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Poul H. Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sharon J. Diskin
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ahmet Sacan
- School of Biomedical Engineering, Science and Health System, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
2
|
Sheridan R, Brennan K, Bazou D, O’Gorman P, Matallanas D, Mc Gee MM. Multiple Myeloma Derived Extracellular Vesicle Uptake by Monocyte Cells Stimulates IL-6 and MMP-9 Secretion and Promotes Cancer Cell Migration and Proliferation. Cancers (Basel) 2024; 16:1011. [PMID: 38473370 PMCID: PMC10930391 DOI: 10.3390/cancers16051011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Multiple Myeloma (MM) is an incurable haematological malignancy caused by uncontrolled growth of plasma cells. MM pathogenesis is attributed to crosstalk between plasma cells and the bone marrow microenvironment, where extracellular vesicles (EVs) play a role. In this study, EVs secreted from a panel of MM cell lines were isolated from conditioned media by ultracentrifugation and fluorescently stained EVs were co-cultured with THP-1 monocyte cells. MM EVs from three cell lines displayed a differential yet dose-dependent uptake by THP-1 cells, with H929 EVs displaying the greatest EV uptake compared to MM.1s and U266 EVs suggesting that uptake efficiency is dependent on the cell line of origin. Furthermore, MM EVs increased the secretion of MMP-9 and IL-6 from monocytes, with H929 EVs inducing the greatest effect, consistent with the greatest uptake efficiency. Moreover, monocyte-conditioned media collected following H929 EV uptake significantly increased the migration and proliferation of MM cells. Finally, EV proteome analysis revealed differential cargo enrichment that correlates with disease progression including a significant enrichment of spliceosome-related proteins in H929 EVs compared to the U266 and MM.1s EVs. Overall, this study demonstrates that MM-derived EVs modulate monocyte function to promote tumour growth and metastasis and reveals possible molecular mechanisms involved.
Collapse
Affiliation(s)
- Rebecca Sheridan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland (K.B.)
| | - Kieran Brennan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland (K.B.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland; (D.B.)
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland; (D.B.)
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland;
| | - Margaret M. Mc Gee
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland (K.B.)
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
3
|
Verbruggen SW, Freeman CL, Freeman FE. Utilizing 3D Models to Unravel the Dynamics of Myeloma Plasma Cells' Escape from the Bone Marrow Microenvironment. Cancers (Basel) 2024; 16:889. [PMID: 38473251 DOI: 10.3390/cancers16050889] [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: 01/25/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Recent therapeutic advancements have markedly increased the survival rates of individuals with multiple myeloma (MM), doubling survival compared to pre-2000 estimates. This progress, driven by highly effective novel agents, suggests a growing population of MM survivors exceeding the 10-year mark post-diagnosis. However, contemporary clinical observations indicate potential trends toward more aggressive relapse phenotypes, characterized by extramedullary disease and dominant proliferative clones, despite these highly effective treatments. To build upon these advances, it is crucial to develop models of MM evolution, particularly focusing on understanding the biological mechanisms behind its development outside the bone marrow. This comprehensive understanding is essential to devising innovative treatment strategies. This review emphasizes the role of 3D models, specifically addressing the bone marrow microenvironment and development of extramedullary sites. It explores the current state-of-the-art in MM modelling, highlighting challenges in replicating the disease's complexity. Recognizing the unique demand for accurate models, the discussion underscores the potential impact of these advanced 3D models on understanding and combating this heterogeneous and still incurable disease.
Collapse
Affiliation(s)
- Stefaan W Verbruggen
- Digital Environment Research Institute, Queen Mary University of London, London E1 4NS, UK
- Center for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK
| | - Ciara L Freeman
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Fiona E Freeman
- School of Mechanical and Materials Engineering, Engineering and Materials Science Centre, University College Dublin, D04 V1W8 Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, D04 V1W8 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Mechanical Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| |
Collapse
|
4
|
Forster S, Radpour R, Ochsenbein AF. Molecular and immunological mechanisms of clonal evolution in multiple myeloma. Front Immunol 2023; 14:1243997. [PMID: 37744361 PMCID: PMC10516567 DOI: 10.3389/fimmu.2023.1243997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by the proliferation of clonal plasma cells in the bone marrow (BM). It is known that early genetic mutations in post-germinal center B/plasma cells are the cause of myelomagenesis. The acquisition of additional chromosomal abnormalities and distinct mutations further promote the outgrowth of malignant plasma cell populations that are resistant to conventional treatments, finally resulting in relapsed and therapy-refractory terminal stages of MM. In addition, myeloma cells are supported by autocrine signaling pathways and the tumor microenvironment (TME), which consists of diverse cell types such as stromal cells, immune cells, and components of the extracellular matrix. The TME provides essential signals and stimuli that induce proliferation and/or prevent apoptosis. In particular, the molecular pathways by which MM cells interact with the TME are crucial for the development of MM. To generate successful therapies and prevent MM recurrence, a thorough understanding of the molecular mechanisms that drive MM progression and therapy resistance is essential. In this review, we summarize key mechanisms that promote myelomagenesis and drive the clonal expansion in the course of MM progression such as autocrine signaling cascades, as well as direct and indirect interactions between the TME and malignant plasma cells. In addition, we highlight drug-resistance mechanisms and emerging therapies that are currently tested in clinical trials to overcome therapy-refractory MM stages.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F. Ochsenbein
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Ishibashi M, Takahashi M, Yamaya T, Imai Y. Current and Future PET Imaging for Multiple Myeloma. Life (Basel) 2023; 13:1701. [PMID: 37629558 PMCID: PMC10455506 DOI: 10.3390/life13081701] [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: 06/07/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Positron emission tomography (PET) is an imaging modality used for the noninvasive assessment of tumor staging and response to therapy. PET with 18F labeled fluorodeoxyglucose (18F-FDG PET) is widely used to assess the active and inactive lesions in patients with multiple myeloma (MM). Despite the availability of 18F-FDG PET for the management of MM, PET imaging is less sensitive than next-generation flow cytometry and sequencing. Therefore, the novel PET radiotracers 64Cu-LLP2A, 68Ga-pentixafor, and 89Zr-daratumumab have been developed to target the cell surface antigens of MM cells. Furthermore, recent studies attempted to visualize the tumor-infiltrating lymphocytes using PET imaging in patients with cancer to investigate their prognostic effect; however, these studies have not yet been performed in MM patients. This review summarizes the recent studies on PET with 18F-FDG and novel radiotracers for the detection of MM and the resulting preclinical research using MM mouse models and clinical studies. Novel PET technologies may be useful for developing therapeutic strategies for MM in the future.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo 113-8602, Japan;
| | - Miwako Takahashi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (M.T.); (T.Y.)
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (M.T.); (T.Y.)
| | - Yoichi Imai
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi 321-0293, Japan
| |
Collapse
|
6
|
Dunphy K, Bazou D, Henry M, Meleady P, Miettinen JJ, Heckman CA, Dowling P, O’Gorman P. Proteomic and Metabolomic Analysis of Bone Marrow and Plasma from Patients with Extramedullary Multiple Myeloma Identifies Distinct Protein and Metabolite Signatures. Cancers (Basel) 2023; 15:3764. [PMID: 37568580 PMCID: PMC10417544 DOI: 10.3390/cancers15153764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Multiple myeloma (MM) is an incurable haematological malignancy of plasma cells in the bone marrow. In rare cases, an aggressive form of MM called extramedullary multiple myeloma (EMM) develops, where myeloma cells enter the bloodstream and colonise distal organs or soft tissues. This variant is associated with refractoriness to conventional therapies and a short overall survival. The molecular mechanisms associated with EMM are not yet fully understood. Here, we analysed the proteome of bone marrow mononuclear cells and blood plasma from eight patients (one serial sample) with EMM and eight patients without extramedullary spread. The patients with EMM had a significantly reduced overall survival with a median survival of 19 months. Label-free mass spectrometry revealed 225 proteins with a significant differential abundance between bone marrow mononuclear cells (BMNCs) isolated from patients with MM and EMM. This plasma proteomics analysis identified 22 proteins with a significant differential abundance. Three proteins, namely vascular cell adhesion molecule 1 (VCAM1), pigment epithelium derived factor (PEDF), and hepatocyte growth factor activator (HGFA), were verified as the promising markers of EMM, with the combined protein panel showing excellent accuracy in distinguishing EMM patients from MM patients. Metabolomic analysis revealed a distinct metabolite signature in EMM patient plasma compared to MM patient plasma. The results provide much needed insight into the phenotypic profile of EMM and in identifying promising plasma-derived markers of EMM that may inform novel drug development strategies.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, Maynooth University, W23 F2K8 Kildare, Ireland;
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 AX57 Dublin, Ireland; (D.B.); (P.O.)
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland; (M.H.); (P.M.)
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland; (M.H.); (P.M.)
| | - Juho J. Miettinen
- Institute for Molecular Medicine Finland-FIMM, HiLIFE–Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland; (J.J.M.); (C.A.H.)
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland-FIMM, HiLIFE–Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland; (J.J.M.); (C.A.H.)
| | - Paul Dowling
- Department of Biology, Maynooth University, W23 F2K8 Kildare, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 AX57 Dublin, Ireland; (D.B.); (P.O.)
| |
Collapse
|
7
|
Khojasteh E, Dehdashti F, Shokeen M. Molecular imaging of bone metastasis. J Bone Oncol 2023; 40:100477. [PMID: 37193117 PMCID: PMC10182320 DOI: 10.1016/j.jbo.2023.100477] [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: 01/30/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 05/18/2023] Open
Abstract
Recent advances in molecularly targeted modular designs for in vivo imaging applications has thrusted open possibilities of investigating deep molecular interactions non-invasively and dynamically. The shifting landscape of biomarker concentration and cellular interactions throughout pathological progression requires quick adaptation of imaging agents and detection modalities for accurate readouts. The synergy of state of art instrumentation with molecularly targeted molecules is resulting in more precise, accurate and reproducible data sets, which is facilitating investigation of several novel questions. Small molecules, peptides, antibodies and nanoparticles are some of the commonly used molecular targeting vectors that can be applied for imaging as well as therapy. The field of theranostics, which encompasses joint application of therapy and imaging, is successfully leveraging the multifunctional use of these biomolecules [[1], [2]]. Sensitive detection of cancerous lesions and accurate assessment of treatment response has been transformative for patient management. Particularly, since bone metastasis is one of the dominant causes of morbidity and mortality in cancer patients, imaging can be hugely impactful in this patient population. The intent of this review is to highlight the utility of molecular positron emission tomography (PET) imaging in the context of prostate and breast bone metastatic cancer, and multiple myeloma. Furthermore, comparisons are drawn with traditionally utilized bone scans (skeletal scintigraphy). Both these modalities can be synergistic or complementary for assessing lytic- and blastic- bone lesions.
Collapse
Affiliation(s)
- Eliana Khojasteh
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Farrokh Dehdashti
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Monica Shokeen
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Corresponding author at: Mallinckrodt Institute of Radiology, 510 South Kingshighway Boulevard, St. Louis, MO 63110, USA.
| |
Collapse
|
8
|
Matamala Montoya M, van Slobbe GJJ, Chang JC, Zaal EA, Berkers CR. Metabolic changes underlying drug resistance in the multiple myeloma tumor microenvironment. Front Oncol 2023; 13:1155621. [PMID: 37091139 PMCID: PMC10117897 DOI: 10.3389/fonc.2023.1155621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells in the bone marrow (BM). MM remains an incurable disease, with the majority of patients experiencing multiple relapses from different drugs. The MM tumor microenvironment (TME) and in particular bone-marrow stromal cells (BMSCs) play a crucial role in the development of drug resistance. Metabolic reprogramming is emerging as a hallmark of cancer that can potentially be exploited for cancer treatment. Recent studies show that metabolism is further adjusted in MM cells during the development of drug resistance. However, little is known about the role of BMSCs in inducing metabolic changes that are associated with drug resistance. In this Perspective, we summarize current knowledge concerning the metabolic reprogramming of MM, with a focus on those changes associated with drug resistance to the proteasome inhibitor Bortezomib (BTZ). In addition, we present proof-of-concept fluxomics (glucose isotope-tracing) and Seahorse data to show that co-culture of MM cells with BMSCs skews the metabolic phenotype of MM cells towards a drug-resistant phenotype, with increased oxidative phosphorylation (OXPHOS), serine synthesis pathway (SSP), TCA cycle and glutathione (GSH) synthesis. Given the crucial role of BMSCs in conveying drug resistance, insights into the metabolic interaction between MM and BMSCs may ultimately aid in the identification of novel metabolic targets that can be exploited for therapy.
Collapse
Affiliation(s)
- María Matamala Montoya
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Gijs J. J. van Slobbe
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jung-Chin Chang
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Esther A. Zaal
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Celia R. Berkers, ; Esther A. Zaal,
| | - Celia R. Berkers
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Celia R. Berkers, ; Esther A. Zaal,
| |
Collapse
|
9
|
Laforest R, Ghai A, Fraum TJ, Oyama R, Frye J, Kaemmerer H, Gaehle G, Voller T, Mpoy C, Rogers BE, Fiala M, Shoghi KI, Achilefu S, Rettig M, Vij R, DiPersio JF, Schwarz S, Shokeen M, Dehdashti F. First-in-Humans Evaluation of Safety and Dosimetry of 64Cu-LLP2A for PET Imaging. J Nucl Med 2023; 64:320-328. [PMID: 36008121 PMCID: PMC9902845 DOI: 10.2967/jnumed.122.264349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 02/04/2023] Open
Abstract
There remains an unmet need for molecularly targeted imaging agents for multiple myeloma (MM). The integrin very late antigen 4 (VLA4), is differentially expressed in malignant MM cells and in pathogenic inflammatory microenvironmental cells. [64Cu]Cu-CB-TE1A1P-LLP2A (64Cu-LLP2A) is a VLA4-targeted, high-affinity radiopharmaceutical with promising utility for managing patients diagnosed with MM. Here, we evaluated the safety and human radiation dosimetry of 64Cu-LLP2A for potential use in MM patients. Methods: A single-dose [natCu]Cu-LLP2A (Cu-LLP2A) tolerability and toxicity study was performed on CD-1 (Hsd:ICR) male and female mice. 64Cu-LLP2A was synthesized in accordance with good-manufacturing-practice-compliant procedures. Three MM patients and six healthy participants underwent 64Cu-LLP2A-PET/CT or PET/MRI at up to 3 time points to help determine tracer biodistribution, pharmacokinetics, and radiation dosimetry. Time-activity curves were plotted for each participant. Mean organ-absorbed doses and effective doses were calculated using the OLINDA software. Tracer bioactivity was evaluated via cell-binding assays, and metabolites from human blood samples were analyzed with analytic radio-high-performance liquid chromatography. When feasible, VLA4 expression was evaluated in the biopsy tissues using 14-color flow cytometry. Results: A 150-fold mass excess of the desired imaging dose was tolerated well in male and female CD-1 mice (no observed adverse effect level). Time-activity curves from human imaging data showed rapid tracer clearance from blood via the kidneys and bladder. The effective dose of 64Cu-LLP2A in humans was 0.036 ± 0.006 mSv/MBq, and the spleen had the highest organ uptake, 0.142 ± 0.034 mSv/MBq. Among all tissues, the red marrow demonstrated the highest residence time. Image quality analysis supports an early imaging time (4-5 h after injection of the radiotracer) as optimal. Cell studies showed statistically significant blocking for the tracer produced for all human studies (82.42% ± 13.47%). Blood metabolism studies confirmed a stable product peak (>90%) up to 1 h after injection of the radiopharmaceutical. No clinical or laboratory adverse events related to 64Cu-LLP2A were observed in the human participants. Conclusion: 64Cu-LLP2A exhibited a favorable dosimetry and safety profile for use in humans.
Collapse
Affiliation(s)
- Richard Laforest
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Anchal Ghai
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Tyler J Fraum
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Reiko Oyama
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer Frye
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Helen Kaemmerer
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Greg Gaehle
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Tom Voller
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Buck E Rogers
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Fiala
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Kooresh I Shoghi
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Samuel Achilefu
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Rettig
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Ravi Vij
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - John F DiPersio
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Sally Schwarz
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Monica Shokeen
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri;
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Farrokh Dehdashti
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri;
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
10
|
Gupta S, Master S, Graham C. Extramedullary Multiple Myeloma: A Patient-Focused Review of the Pathogenesis of Bone Marrow Escape. World J Oncol 2022; 13:311-319. [PMID: 36406195 PMCID: PMC9635794 DOI: 10.14740/wjon1521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/22/2022] [Indexed: 08/11/2023] Open
Abstract
Multiple myeloma (MM) is a neoplastic clonal proliferation of plasma cells, predominantly in the bone marrow. The presentation of MM in extramedullary tissue, particularly the liver, is uncommon with only a few reported cases in literature. We report a rare and unusual presentation of kappa light chain restricted MM with progression of disease to involve the liver. MM was initially diagnosed on bone marrow biopsy, initially treated with carfilzomib, lenalidomide and dexamethasone, later changed to bortezomib, daratumumab and dexamethasone. There was subsequent progression with a new biopsy-proven myelomatous liver lesion. The patient could not receive high-dose chemotherapy due to multiple co-morbidities and extent of disease and eventually succumbed to her disease rapidly. This article emphasizes the poor prognosis of extramedullary involvement in MM and the pathogenic mechanisms by which it develops. Based on a review of the literature of other cases and case series of solitary or diffuse myeloma involvement in the liver, high-dose chemotherapy in combination with proteasome inhibitors and immunomodulators has the best success rate with less relapse and progressive disease in extramedullary myeloma. Our analysis concluded that the gain of CD44, loss of CD56, loss of very late antigen-4 (VLA-4), imbalance of the chemokine receptor-4-chemokine ligand-12 (CXCR4-CXCL12) axis, metastasis-associated lung adenocarcinoma 1 (MALAT1) upregulation, RAS pathway activation as well as 13q and 17p deletions show an increased propensity of malignant plasma cells to leave the bone marrow and hone in extramedullary sites giving rise to more aggressive extramedullary diseases. Targeted therapeutics such as CD44v-directed therapy and reactivation of p53 to wild-type conformation could potentially be evaluated as treatment options in the future to improve outcomes in this aggressive form of MM, especially in patients with advanced disease and limited treatment options.
Collapse
Affiliation(s)
- Supriya Gupta
- Division of Hematology-Oncology, Feist Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Samip Master
- Division of Hematology-Oncology, Feist Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher Graham
- Division of Hematology-Oncology, Feist Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
11
|
Holstein SA, Asimakopoulos F, Azab AK, Bianchi G, Bhutani M, Crews LA, Cupedo T, Giles H, Gooding S, Hillengass J, John L, Kaiser S, Lee L, Maclachlan K, Pasquini MC, Pichiorri F, Shah N, Shokeen M, Shy BR, Smith EL, Verona R, Usmani SZ, McCarthy PL. Proceedings from the Blood and Marrow Transplant Clinical Trials Network Myeloma Intergroup Workshop on Immune and Cellular Therapy in Multiple Myeloma. Transplant Cell Ther 2022; 28:446-454. [PMID: 35605882 PMCID: PMC9357156 DOI: 10.1016/j.jtct.2022.05.019] [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: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022]
Abstract
The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) Myeloma Intergroup conducted a workshop on Immune and Cellular Therapy in Multiple Myeloma on January 7, 2022. This workshop included presentations by basic, translational, and clinical researchers with expertise in plasma cell dyscrasias. Four main topics were discussed: platforms for myeloma disease evaluation, insights into pathophysiology, therapeutic target and resistance mechanisms, and cellular therapy for multiple myeloma. Here we provide a comprehensive summary of these workshop presentations.
Collapse
Affiliation(s)
| | - Fotis Asimakopoulos
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | | | - Giada Bianchi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Leslie A Crews
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Tom Cupedo
- ErasmusMC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Hannah Giles
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Sarah Gooding
- MRC Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Lukas John
- University Hospital Heidelberg, Heidelberg, Germany
| | | | - Lydia Lee
- University College London, London, United Kingdom
| | | | | | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California; Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, California
| | - Nina Shah
- University of California San Francisco, San Francisco, California
| | - Monica Shokeen
- Washington University School of Medicine, St. Louis, Missouri
| | - Brian R Shy
- University of California San Francisco, San Francisco, California
| | - Eric L Smith
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Raluca Verona
- Janssen Research & Development, Spring House, Pennsylvania
| | - Saad Z Usmani
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
12
|
Forster S, Radpour R. Molecular Impact of the Tumor Microenvironment on Multiple Myeloma Dissemination and Extramedullary Disease. Front Oncol 2022; 12:941437. [PMID: 35847862 PMCID: PMC9284036 DOI: 10.3389/fonc.2022.941437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/06/2022] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the most common malignant monoclonal disease of plasma cells. Aside from classical chemotherapy and glucocorticoids, proteasome inhibitors, immunomodulatory agents and monoclonal antibodies are used in the current treatment scheme of MM. The tumor microenvironment (TME) plays a fundamental role in the development and progression of numerous solid and non-solid cancer entities. In MM, the survival and expansion of malignant plasma cell clones heavily depends on various direct and indirect signaling pathways provided by the surrounding bone marrow (BM) niche. In a number of MM patients, single plasma cell clones lose their BM dependency and are capable to engraft at distant body sites or organs. The resulting condition is defined as an extramedullary myeloma (EMM). EMMs are highly aggressive disease stages linked to a dismal prognosis. Emerging literature demonstrates that the dynamic interactions between the TME and malignant plasma cells affect myeloma dissemination. In this review, we aim to summarize how the cellular and non-cellular BM compartments can promote plasma cells to exit their BM niche and metastasize to distant intra-or extramedullary locations. In addition, we list selected therapy concepts that directly target the TME with the potential to prevent myeloma spread.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- *Correspondence: Ramin Radpour,
| |
Collapse
|
13
|
Janicka N, Sałek A, Sawińska M, Kuchar E, Wiela-Hojeńska A, Karłowicz-Bodalska K. Effects of Non-Opioid Analgesics on the Cell Membrane of Skin and Gastrointestinal Cancers. Int J Mol Sci 2022; 23:ijms23137096. [PMID: 35806101 PMCID: PMC9266389 DOI: 10.3390/ijms23137096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
Skin and gastrointestinal cancer cells are the target of research by many scientists due to the increasing morbidity and mortality rates around the world. New indications for drugs used in various conditions are being discovered. Non-opioid analgesics are worth noting as very popular, widely available, relatively cheap medications. They also have the ability to modulate the membrane components of tumor cells. The aim of this review is to analyze the impact of diclofenac, ibuprofen, naproxen, acetylsalicylic acid and paracetamol on skin and gastrointestinal cancers cell membrane. These drugs may affect the membrane through topical application, at the in vitro and in vivo level after oral or parenteral administration. They can lead to up- or downregulated expression of receptors, transporters and other molecules associated with plasma membrane. Medications may also alter the lipid bilayer composition of membrane, resulting in changes in its integrity and fluidity. Described modulations can cause the visualization of cancer cells, enhanced response of the immune system and the initiation of cell death. The outcome of this is inhibition of progression or reduction of tumor mass and supports chemotherapy. In conclusion, non-opioid analgesics may be used in the future as adjunctive therapy for the treatment of these cancers.
Collapse
Affiliation(s)
- Natalia Janicka
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (N.J.); (A.S.); (M.S.)
| | - Agnieszka Sałek
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (N.J.); (A.S.); (M.S.)
| | - Magdalena Sawińska
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (N.J.); (A.S.); (M.S.)
| | - Ernest Kuchar
- Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Anna Wiela-Hojeńska
- Department of Clinical Pharmacology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Katarzyna Karłowicz-Bodalska
- Department of Drugs Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Correspondence:
| |
Collapse
|