1
|
Mahalingam D, Owonikoko TK, Delpassand E, Mulcahy MF, Kalyan A, Ulahannan S, Cheung K, Izumi Y, Johansen M, Madden T, Shimoyama S, Subach RA, Suzuki T, Wages DS, Wheeler C, Richardson DL. A trial of radiolabeled antibody yttrium-90-FF-21101 for the treatment of advanced ovarian and other cancers. Cancer 2025; 131:e35680. [PMID: 39748726 PMCID: PMC11696210 DOI: 10.1002/cncr.35680] [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: 06/19/2024] [Revised: 10/21/2024] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Yttrium-90 FF-21101 (90Y-FF-21101) is a radiopharmaceutical that targets P-cadherin as a therapy against solid tumors. A previously reported, first-in-human study determined that a dose of 25 mCi/m2 was safe, and a patient with clear cell carcinoma of the ovary achieved a complete response. In this article, the authors report the results of 90Y-FF-21101 treatment in an ovarian carcinoma expansion cohort and in patients with selected solid tumors who had known high P-cadherin expression. METHODS The trial was conducted as an open-label study in patients with advanced/metastatic disease. Radiologic response and safety were evaluated in patients who received 25 mCi/m2 intravenously once every three cycles of 28 days until they developed progressive disease. Evaluation of the ovarian cohort was conducted in a Simon two-stage manner to determine further enrollment. RESULTS Fifty-seven patients (20 with ovarian carcinoma) were enrolled and treated. Patients who had ovarian and solid tumors had received a median of five and three prior therapies, respectively. No complete or partial responses were observed, so the trial was ended. The median progression-free survival was 118 days for the ovarian cohort and 55 days for the solid-tumor cohort. The most common treatment-related adverse events were thrombocytopenia (40%) and neutropenia (54%). One patient each developed fatal veno-occlusive disease and intracranial hemorrhage. Patients with higher P-cadherin levels remained on the study longer. CONCLUSIONS 90Y-FF-21101 did not meet the predefined efficacy criteria, and adverse events were consistent with 90Y agents. These data may assist in the development of other P-cadherin-directed therapies (ClinicalTrials.gov identifier NCT02454010).
Collapse
Affiliation(s)
| | - Taofeek K. Owonikoko
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
- Present address:
AbbVie GKTokyoJapan
| | | | - Mary F. Mulcahy
- Northwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Aparna Kalyan
- Northwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Susanna Ulahannan
- Stephenson Cancer CenterUniversity of Oklahoma Health Sciences Center/Sarah Cannon Research InstituteOklahoma CityOklahomaUSA
| | - Kin Cheung
- FUJIFILM Pharmaceuticals USA, IncCambridgeMassachusettsUSA
| | - Yasayuki Izumi
- FUJIFILM Pharmaceuticals USA, IncCambridgeMassachusettsUSA
| | - Mary Johansen
- FUJIFILM Pharmaceuticals USA, IncCambridgeMassachusettsUSA
| | - Timothy Madden
- FUJIFILM Pharmaceuticals USA, IncCambridgeMassachusettsUSA
| | | | | | - Takeaki Suzuki
- FUJIFILM Pharmaceuticals USA, IncCambridgeMassachusettsUSA
| | - David S. Wages
- FUJIFILM Pharmaceuticals USA, IncCambridgeMassachusettsUSA
- Present address:
Deln3 Consulting LLCSouthboroughMAUSA
| | | | - Debra L. Richardson
- Stephenson Cancer CenterUniversity of Oklahoma Health Sciences Center/Sarah Cannon Research InstituteOklahoma CityOklahomaUSA
| |
Collapse
|
2
|
Roy I, Krishnan S, Kabashin AV, Zavestovskaya IN, Prasad PN. Transforming Nuclear Medicine with Nanoradiopharmaceuticals. ACS NANO 2022; 16:5036-5061. [PMID: 35294165 DOI: 10.1021/acsnano.1c10550] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nuclear medicine is expected to make major advances in cancer diagnosis and therapy; tumor-targeted radiopharmaceuticals preferentially eradicate tumors while causing minimal damage to healthy tissues. The current scope of nuclear medicine can be significantly expanded by integration with nanomedicine, which utilizes nanoparticles for cancer diagnosis and therapy by capitalizing on the increased surface area-to-volume ratio, the passive/active targeting ability and high loading capacity, the greater interaction cross section with biological tissues, the rich surface properties of nanomaterials, the facile decoration of nanomaterials with a plethora of functionalities, and the potential for multiplexing several functionalities within one construct. This review provides a comprehensive discussion of nuclear nanomedicine using tumor-targeted nanoparticles for cancer radiation therapy with either pre-embedded radionuclides or nonradioactive materials which can be extrinsically triggered using various external nuclear particle sources to produce in situ radioactivity. In addition, it describes the prospect of combining nuclear nanomedicine with other modalities to enable synergistically enhanced combination therapies. The review also discusses advances in the fabrication of radionuclides as well as describes laser ablation technologies for producing nanoradiopharmaceuticals, which combine the ease of production with exceptional purity and rapid biodegradability, along with additional imaging or therapeutic functionalities. From a practical standpoint, these attributes of nanoradiopharmaceuticals may provide distinct advantages in diagnostic/therapeutic sensitivity and specificity, imaging resolution, and scalability of turnkey platforms. Coupling image-guided targeted radiation therapy with the possibility of in situ activation of nanomaterials as well as combining with other therapeutic modalities using a multifunctional nanoplatform could herald an era of exciting technological and therapeutic advances to radically transform the landscape of nuclear medicine. The review concludes with a discussion of current challenges and presents the authors' views on future opportunities to stimulate further research in this rewarding field of high societal impact.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy - Case 917, 13288 Marseille, France
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
| | - Irina N Zavestovskaya
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Nuclear Physics and Astrophysics Department, LPI of RAS, 119991 Moscow, Russia
| | - Paras N Prasad
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Department of Chemistry and Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
3
|
Nautiyal A, Jha AK, Mithun S, Shetye B, Kameswaran M, Shah S, Rangarajan V, Gupta S. Analysis of absorbed dose in radioimmunotherapy with 177Lu-trastuzumab using two different imaging scenarios: a pilot study. Nucl Med Commun 2021; 42:1382-1395. [PMID: 34406146 DOI: 10.1097/mnm.0000000000001472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Internal organ dosimetry is an important procedure to demonstrate the reliable application of 177Lu-trastuzumab radioimmunotherapy for human epidermal growth factor receptor-positive metastatic breast cancers. We are reporting the first human dosimetry study for 177Lu-trastuzumab. Another objective of our study was to calculate and compare the absorbed doses for normal organs and tumor lesions in patients before radioimmunotherapy with 177Lu-trastuzumab using two different imaging scenarios. METHODS Eleven patients (48.27 ± 8.95 years) with a history of metastatic breast cancer were included in the study. Postadministration of 177Lu-trastuzumab (351.09 ± 23.89 MBq/2 mg), acquisition was performed using planar and hybrid imaging scenarios at 4, 24, 72 and 168 h. Single-photon emission computed tomography/computed tomography imaging was performed at 72 h postinjection. Acquired images were processed using Dosimetry Toolkit software for the estimation of normalized cumulated activity in organs and tumor lesions. OLINDA/EXM 2.0 software was used for absorbed dose calculation in both scenarios. RESULTS Significant difference in normalized cumulated activity and the absorbed dose is noted between two imaging scenarios for the organs and tumor lesions (P < 0.05). Mean absorbed dose (mGy/MBq) estimated from heart, lungs, liver, spleen, kidney, adrenal, pancreas and colon using planar and hybrid scenarios were 0.81 ± 0.19 and 0.63 ± 0.17; 0.75 ± 0.13 and 0.32 ± 0.06; 1.26 ± 0.25 and 1.01 ± 0.17; 0.68 ± 0.22 and 0.53 ± 0.16; 0.91 ± 0.3 and 0.69 ± 0.24; 0.18 ± 0.04 and 0.11 ± 0.02; 0.25 ± 0.22 and 0.09 ± 0.02 and 0.75 ± 0.61 and 0.44 ± 0.28, respectively. CONCLUSIONS On the basis of our dosimetric evaluation, we concluded that radioimmunotherapy with 177Lu-trastuzumab is well tolerated to be implemented in routine clinical practice against HER2 positive metastatic breast cancer. Liver is the main critical organ at risk. Hybrid scenario demonstrated significantly lower absorbed doses in organs and tumors compared to the multiplanar method.
Collapse
Affiliation(s)
- Amit Nautiyal
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ashish K Jha
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sneha Mithun
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Bhakti Shetye
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mythili Kameswaran
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sneha Shah
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Centre, Parel
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sudeep Gupta
- Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC)
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
4
|
Rahumatullah A, Lim TS, Yunus MH, Noordin R. Development of an Antigen Detection ELISA for Bancroftian Filariasis Using BmSXP-Specific Recombinant Monoclonal Antibody. Am J Trop Med Hyg 2020; 101:436-440. [PMID: 31162018 DOI: 10.4269/ajtmh.19-0034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lymphatic filariasis is a mosquito-borne parasitic disease responsible for morbidity and disability that affects 1.2 billion people worldwide, mainly the poor communities. Currently, filarial antigen testing is the method of choice for the detection of bancroftian filariasis, and to date, there are two commonly used tests. In the present study, a recently reported recombinant monoclonal antibody (5B) specific to BmSXP filarial antigen was used in developing an ELISA for the detection of circulating filarial antigen in sera of patients with bancroftian filariasis. The performance of the ELISA was evaluated using 124 serum samples. The ELISA was positive with all sera from microfilaremic bancroftian filariasis patients (n = 34). It also showed 100% diagnostic specificity when tested with sera from 50 healthy individuals and 40 patients with other parasitic diseases. The developed assay using the novel 5B recombinant monoclonal antibody could potentially be a promising alternative antigen detection test for bancroftian filariasis.
Collapse
Affiliation(s)
- Anizah Rahumatullah
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Muhammad Hafiznur Yunus
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Rahmah Noordin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Pulau Pinang, Malaysia
| |
Collapse
|
5
|
Shek D, Read SA, Ahlenstiel G, Piatkov I. Pharmacogenetics of anticancer monoclonal antibodies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:69-81. [PMID: 35582142 PMCID: PMC9019180 DOI: 10.20517/cdr.2018.20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/09/2019] [Accepted: 02/19/2019] [Indexed: 12/16/2022]
Abstract
Pharmacogenetics is the study of therapeutic and adverse responses to drugs based on an individual’s genetic background. Monoclonal antibodies (mAbs) are a rapidly evolving field in cancer therapy, however a number of newly developed and highly effective mAbs (e.g., anti-CTLA-4 and anti-PD-1) possess pharmacogenomic profiles that remain largely undefined. Since the first chemotherapeutic mAb Rituximab was approved in 1997 by the US Food and Drug Administration for cancer treatment, a broad number of other mAbs have been successfully developed and implemented into oncological practice. Nowadays, mAbs are considered as one of the most promising new approaches for cancer treatment. The efficacy of mAb treatment can however be significantly affected by genetic background, where genes responsible for antibody presentation and metabolism, for example, can seriously affect patient outcome. This review will focus on current anticancer mAb treatments, patient genetics that shape their efficacy, and the molecular pathways that bridge the two.
Collapse
Affiliation(s)
- Dmitrii Shek
- Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia
| | - Scott A Read
- Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia.,Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Golo Ahlenstiel
- Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia.,Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia.,Blacktown Hospital, Blacktown, NSW 2148, Australia
| | | |
Collapse
|