Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.390
Filter
1.
Proc Natl Acad Sci U S A ; 121(28): e2322203121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968122

ABSTRACT

Targeting cell surface molecules using radioligand and antibody-based therapies has yielded considerable success across cancers. However, it remains unclear how the expression of putative lineage markers, particularly cell surface molecules, varies in the process of lineage plasticity, wherein tumor cells alter their identity and acquire new oncogenic properties. A notable example of lineage plasticity is the transformation of prostate adenocarcinoma (PRAD) to neuroendocrine prostate cancer (NEPC)-a growing resistance mechanism that results in the loss of responsiveness to androgen blockade and portends dismal patient survival. To understand how lineage markers vary across the evolution of lineage plasticity in prostate cancer, we applied single-cell analyses to 21 human prostate tumor biopsies and two genetically engineered mouse models, together with tissue microarray analysis on 131 tumor samples. Not only did we observe a higher degree of phenotypic heterogeneity in castrate-resistant PRAD and NEPC than previously anticipated but also found that the expression of molecules targeted therapeutically, namely PSMA, STEAP1, STEAP2, TROP2, CEACAM5, and DLL3, varied within a subset of gene-regulatory networks (GRNs). We also noted that NEPC and small cell lung cancer subtypes shared a set of GRNs, indicative of conserved biologic pathways that may be exploited therapeutically across tumor types. While this extreme level of transcriptional heterogeneity, particularly in cell surface marker expression, may mitigate the durability of clinical responses to current and future antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.


Subject(s)
Single-Cell Analysis , Male , Humans , Single-Cell Analysis/methods , Animals , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Antigens, Surface/metabolism , Antigens, Surface/genetics , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/drug therapy , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/drug therapy , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy
2.
Theranostics ; 14(9): 3623-3633, 2024.
Article in English | MEDLINE | ID: mdl-38948055

ABSTRACT

Introduction: Prostate Specific Membrane Antigen Positron Emission Tomography (PSMA-PET) is routinely used for the staging of patients with prostate cancer, but data on response assessment are sparse and primarily stem from metastatic castration-resistant prostate cancer (mCRPC) patients treated with PSMA radioligand therapy. Still, follow-up PSMA-PET is employed in earlier disease stages in case of clinical suspicion of disease persistence, recurrence or progression to decide if localized or systemic treatment is indicated. Therefore, the prognostic value of PSMA-PET derived tumor volumes in earlier disease stages (i.e., hormone-sensitive prostate cancer (HSPC) and non-[177Lu]Lu-PSMA-617 (LuPSMA) therapy castration resistant prostate cancer (CRPC)) are evaluated in this manuscript. Methods: A total number of 73 patients (6 primary staging, 42 HSPC, 25 CRPC) underwent two (i.e., baseline and follow-up, median interval: 379 days) whole-body [68Ga]Ga-PSMA-11 PET/CT scans between Nov 2014 and Dec 2018. Analysis was restricted to non-LuPSMA therapy patients. PSMA-PETs were retrospectively analyzed and primary tumor, lymph node-, visceral-, and bone metastases were segmented. Body weight-adjusted organ-specific and total tumor volumes (PSMAvol: sum of PET volumes of all lesions) were measured for baseline and follow-up. PSMAvol response was calculated as the absolute difference of whole-body tumor volumes. High metastatic burden (>5 metastases), RECIP 1.0 and PSMA-PET Progression Criteria (PPP) were determined. Survival data were sourced from the cancer registry. Results: The average number of tumor lesions per patient on the initial PET examination was 10.3 (SD 28.4). At baseline, PSMAvol was strongly associated with OS (HR 3.92, p <0.001; n = 73). Likewise, response in PSMAvol was significantly associated with OS (HR 10.48, p < 0.005; n = 73). PPP achieved significance as well (HR 2.19, p <0.05, n = 73). Patients with hormone sensitive disease and poor PSMAvol response (upper quartile of PSMAvol change) in follow-up had shorter outcome (p < 0.05; n = 42). PSMAvol in bones was the most relevant parameter for OS prognostication at baseline and for response assessment (HR 31.11 p < 0.001; HR 32.27, p < 0.001; n = 73). Conclusion: PPP and response in PSMAvol were significantly associated with OS in the present heterogeneous cohort. Bone tumor volume was the relevant miTNM region for OS prognostication. Future prospective evaluation of the performance of organ specific PSMAvol in more homogeneous cohorts seems warranted.


Subject(s)
Positron Emission Tomography Computed Tomography , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Aged , Positron Emission Tomography Computed Tomography/methods , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/pathology , Middle Aged , Follow-Up Studies , Gallium Radioisotopes , Retrospective Studies , Aged, 80 and over , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Glutamate Carboxypeptidase II/metabolism , Radiopharmaceuticals , Antigens, Surface/metabolism , Gallium Isotopes , Prognosis , Lutetium/therapeutic use , Positron-Emission Tomography/methods , Tumor Burden , Heterocyclic Compounds, 1-Ring/therapeutic use , Dipeptides/therapeutic use
3.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928224

ABSTRACT

Breast cancer, known for its diverse subtypes, ranks as one of the leading causes of cancer-related deaths. Prostate-specific membrane antigen (PSMA), primarily associated with prostate cancer, has also been identified in breast cancer, though its role remains unclear. This study aimed to evaluate PSMA expression across different subtypes of early-stage breast cancer and investigate its correlation with clinicopathological factors. This retrospective study included 98 breast cancer cases. PSMA expression was examined in both tumor cells and tumor-associated blood vessels. The analysis revealed PSMA expression in tumor-associated blood vessels in 88 cases and in tumor cells in 75 cases. Ki67 expression correlated positively with PSMA expression in blood vessels (p < 0.0001, RSpearman 0.42) and tumor cells (p = 0.010, RSpearman 0.26). The estrogen and progesterone receptor expression correlated negatively with PSMA levels in blood vessels (p = 0.0053, R Spearman -0.26 and p = 0.00026, R Spearman -0.347, respectively). Human epidermal growth factor receptor 2 (HER2) status did not significantly impact PSMA expression. We did not detect any statistically significant differences between breast cancer subtypes. These findings provide evidence for a heterogenous PSMA expression in breast cancer tissue and suggest its correlation with tumor aggressiveness. Despite the limited sample size, the study provides valuable insights into the potential of PSMA as a prognostic, diagnostic, and therapeutic target in the management of breast cancer.


Subject(s)
Antigens, Surface , Biomarkers, Tumor , Breast Neoplasms , Glutamate Carboxypeptidase II , Immunohistochemistry , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Glutamate Carboxypeptidase II/metabolism , Middle Aged , Antigens, Surface/metabolism , Aged , Biomarkers, Tumor/metabolism , Retrospective Studies , Neoplasm Staging , Adult , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Aged, 80 and over
4.
Mol Pharm ; 21(7): 3256-3267, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856975

ABSTRACT

Prostate-specific membrane antigen (PSMA) overexpressed in prostate cancer cells can serve as a target for imaging and radioligand therapy (RLT). Previously, [68Ga]Ga-P16-093, containing a Ga(III) chelator, N,N'-bis[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid (HBED-CC), displayed excellent PSMA-targeting properties and showed a high tumor uptake and retention useful for diagnosis in prostate cancer patients. Recently, [177Lu]Lu-PSMA-617 has been approved by the U.S. food and drug administration (FDA) for the treatment of prostate cancer patients. Derivatives of PSMA-093 using AAZTA (6-amino-6-methylperhydro-1,4-diazepinetetraacetic acid), as the chelator, were designed as alternative agents forming complexes with both diagnostic and therapeutic radiometals, such as gallium-68 (log K = 22.18) or lutetium-177 (log K = 21.85). The aim of this study is to evaluate AAZTA-Gly-O-(methylcarboxy)-Tyr-Phe-Lys-NH-CO-NH-Glu (designated as AZ-093, 1) leading to a gallium-68/lutetium-177 theranostic pair as potential PSMA targeting agents. Synthesis of the desired precursor, AZ-093, 1, was effectively accomplished. Labeling with either [68Ga]GaCl3 or [177Lu]LuCl3 in a sodium acetate buffer solution (pH 4-5) at 50 °C in 5 to 15 min produced either [68Ga]Ga-1 or [177Lu]Lu-1 with high yields and excellent radiochemical purities. Results of in vitro binding studies, cell uptake, and retention (using PSMA-positive prostate carcinoma cells line, 22Rv1-FOLH1-oe) were comparable to that of [68Ga]Ga-P16-093 and [177Lu]Lu-PSMA-617, respectively. Specific cellular uptake was determined with or without the competitive blocking agent (2 µM of "cold" PSMA-11). Cellular binding and internalization showed a time-dependent increase over 2 h at 37 °C in the PSMA-positive cells. The cell uptakes were completely blocked by the "cold" PSMA-11 suggesting that they are competing for the same PSMA binding sites. In the mouse model with implanted PSMA-positive tumor cells, both [68Ga]Ga-1 and [177Lu]Lu-1 displayed excellent uptake and retention in the tumor. Results indicate that [68Ga]Ga/[177Lu]Lu-1 (68Ga]Ga/[177Lu]Lu-AZ-093) is potentially useful as PSMA-targeting agent for both diagnosis and radiotherapy of prostate cancer.


Subject(s)
Antigens, Surface , Gallium Radioisotopes , Glutamate Carboxypeptidase II , Lutetium , Prostatic Neoplasms , Radiopharmaceuticals , Male , Humans , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/metabolism , Lutetium/chemistry , Antigens, Surface/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/pharmacokinetics , Glutamate Carboxypeptidase II/metabolism , Glutamate Carboxypeptidase II/antagonists & inhibitors , Cell Line, Tumor , Radioisotopes/chemistry , Animals , Chelating Agents/chemistry , Prostate-Specific Antigen/metabolism , Tissue Distribution , Mice , Edetic Acid/analogs & derivatives , Edetic Acid/chemistry , Positron Emission Tomography Computed Tomography/methods
5.
J Nucl Med ; 65(7): 1087-1094, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38844360

ABSTRACT

Benchtop 99Mo/99mTc and 188W/188Re generators enable economical production of molecular theranostic 99mTc and 188Re radiopharmaceuticals, provided that simple, kit-based chemistry exists to radiolabel targeting vectors with these radionuclides. We have previously described a diphosphine platform that efficiently incorporates 99mTc into receptor-targeted peptides. Here, we report its application to label a prostate-specific membrane antigen (PSMA)-targeted peptide with 99mTc and 188Re for diagnostic imaging and systemic radiotherapy of prostate cancer. Methods: Two diphosphine-dipeptide bioconjugates, DP1-PSMAt and DP2-PSMAt, were formulated into kits for radiolabeling with 99mTc and 188Re. The resulting radiotracers were studied in vitro, in prostate cancer cells, and in vivo in mouse xenograft models, to assess similarity of uptake and biodistribution for each 99mTc/188Re pair of agents. Results: Both DP1-PSMAt and DP2-PSMAt could be efficiently radiolabeled with 99mTc and 188Re using kit-based methods to furnish the isostructural compounds M-DP1-PSMAt and M-DP2-PSMAt (M = [99mTc]Tc, [188Re]Re). All 99mTc/188Re radiotracers demonstrated specific uptake in PSMA-expressing prostate cancer cells, with negligible uptake in prostate cancer cells that did not express PSMA or in which PSMA uptake was blocked. M-DP1-PSMAt and M-DP2-PSMAt also exhibited high tumor uptake (18-30 percentage injected dose per gram at 2 h after injection), low retention in nontarget organs, fast blood clearance, and excretion predominantly via a renal pathway. Importantly, each pair of 99mTc/188Re radiotracers showed near-identical biologic behavior in these experiments. Conclusion: We have prepared and developed novel pairs of isostructural PSMA-targeting 99mTc/188Re theranostic agents. These generator-based theranostic agents have potential to provide access to the benefits of PSMA-targeted diagnostic imaging and systemic radiotherapy in health care settings that do not routinely have access to either reactor-produced 177Lu radiopharmaceuticals or PET/CT infrastructure.


Subject(s)
Prostatic Neoplasms , Radioisotopes , Rhenium , Technetium , Male , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/metabolism , Mice , Rhenium/chemistry , Animals , Humans , Technetium/chemistry , Radioisotopes/chemistry , Cell Line, Tumor , Tissue Distribution , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Theranostic Nanomedicine , Peptides/chemistry , Precision Medicine
6.
ACS Nano ; 18(26): 16674-16683, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38907991

ABSTRACT

Targeted nanoparticles have been extensively explored for their ability to deliver their payload to a selective cell population while reducing off-target side effects. The design of actively targeted nanoparticles requires the grafting of a ligand that specifically binds to a highly expressed receptor on the surface of the targeted cell population. Optimizing the interactions between the targeting ligand and the receptor can maximize the cellular uptake of the nanoparticles and subsequently improve their activity. Here, we evaluated how the density and presentation of the targeting ligands dictate the cellular uptake of nanoparticles. To do so, we used a DNA-scaffolded PLGA nanoparticle system to achieve efficient and tunable ligand conjugation. A prostate-specific membrane antigen (PSMA) expressing a prostate cancer cell line was used as a model. The density and presentation of PSMA targeting ligand ACUPA were precisely tuned on the DNA-scaffolded nanoparticle surface, and their impact on cellular uptake was evaluated. It was found that matching the ligand density with the cell receptor density achieved the maximum cellular uptake and specificity. Furthermore, DNA hybridization-mediated targeting chain rigidity of the DNA-scaffolded nanoparticle offered ∼3 times higher cellular uptake compared to the ACUPA-terminated PLGA nanoparticle. Our findings also indicated a ∼ 3.7-fold reduction in the cellular uptake for the DNA hybridization of the non-targeting chain. We showed that nanoparticle uptake is energy-dependent and follows a clathrin-mediated pathway. Finally, we validated the preferential tumor targeting of the nanoparticles in a bilateral tumor xenograft model. Our results provide a rational guideline for designing actively targeted nanoparticles and highlight the application of DNA-scaffolded nanoparticles as an efficient active targeting platform.


Subject(s)
DNA , Glutamate Carboxypeptidase II , Nanoparticles , Prostatic Neoplasms , Nanoparticles/chemistry , Humans , DNA/chemistry , DNA/metabolism , Ligands , Male , Glutamate Carboxypeptidase II/metabolism , Glutamate Carboxypeptidase II/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Mice , Antigens, Surface/metabolism , Antigens, Surface/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
7.
Clin Nucl Med ; 49(8): e394-e395, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38847784

ABSTRACT

ABSTRACT: Although PSMA-targeted PET imaging is predominantly used for prostate carcinoma (PC), it has also been reported for thyroid carcinoma (TC). A 77-year-old man had a liver metastasectomy for poorly differentiated TC, which had elevated 18 F-FDG uptake. Two years later, he was diagnosed with acinar-type modified Gleason score of 7 (3 + 4) PC. Four years later, he had metastatic liver lesions that had no radioactive iodine and 18 F-FDG avidity. These lesions were 68 Ga-PSMA avid, and the biopsy confirmed TC metastasis. This case emphasizes the importance of 68 Ga-PSMA-based imaging in poorly differentiated TC and pathological confirmation for lesions that were 68 Ga-PSMA-positive.


Subject(s)
Liver Neoplasms , Prostatic Neoplasms , Thyroid Neoplasms , Humans , Male , Aged , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Edetic Acid/analogs & derivatives , Oligopeptides , Gallium Radioisotopes , Gallium Isotopes , Positron Emission Tomography Computed Tomography , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism
8.
Clin Nucl Med ; 49(8): e383-e389, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38847441

ABSTRACT

PURPOSE: We investigated the impact of prostate-specific membrane antigen (PSMA) PET/CT compared with conventional imaging on treatment outcomes for node-positive prostate cancer (PCa) patients who underwent androgen deprivation therapy (ADT) and external radiotherapy (RT). PATIENTS AND METHODS: A multicentric, retrospective study recruited patients with node-positive PCa patients who underwent conventional radiological evaluation or PSMA PET/CT and received ADT and RT at 3 hospitals from 2009 to 2021 were enrolled. Patients underwent prostate and pelvis RT, accompanied by a minimum of 6 months of ADT. The primary endpoints were progression-free survival (PFS) and PCa-specific survival (PCSS). Cox regression analyzed the association of survival with potential prognostic factors, whereas logistic regression identified the predictors of bone and lymph node metastasis. RESULTS: The median follow-up time was 64.0 months. The majority of patients (64.1%) underwent PSMA PET/CT for staging. The 5-year rates of PFS and PCSS were 63.7% and 83.7%, respectively. Disease progression was observed in 90 patients (36.3%). In multivariable analysis, ADT duration of less than 24 months and post-RT prostate-specific antigen (PSA) nadir were prognostic for PFS. Early clinical T stage and PSMA PET/CT predicted better PCSS. Patients staged with PSMA PET/CT had exhibited significantly higher 5-year PCSS rates than compared with those staged with conventional imaging (95.1% vs 76.9%; P = 0.01). Shorter ADT duration and higher PSA levels after RT independently predicted bone metastasis in multivariable logistic regression. Advanced T stage, shorter ADT duration, and higher PSA levels after neoadjuvant ADT predicted nonregional lymph node recurrence. CONCLUSIONS: ADT with pelvis RT is an effective treatment option for node-positive PCa patients. The PSMA PET/CT outperformed conventional imaging in PCSS, emphasizing the importance of precise clinical staging for patients undergoing definitive RT.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Lymphatic Metastasis , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/drug therapy , Aged , Retrospective Studies , Treatment Outcome , Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Middle Aged , Aged, 80 and over
9.
Theranostics ; 14(8): 3043-3079, 2024.
Article in English | MEDLINE | ID: mdl-38855174

ABSTRACT

In 1853, the perception of prostate cancer (PCa) as a rare ailment prevailed, was described by the eminent Londoner surgeon John Adams. Rapidly forward to 2018, the landscape dramatically altered. Currently, men face a one-in-nine lifetime risk of PCa, accentuated by improved diagnostic methods and an ageing population. With more than three million men in the United States alone grappling with this disease, the overall risk of succumbing to stands at one in 39. The intricate clinical and biological diversity of PCa poses serious challenges in terms of imaging, ongoing monitoring, and disease management. In the field of theranostics, diagnostic and therapeutic approaches that harmoniously merge targeted imaging with treatments are integrated. A pivotal player in this arena is radiotheranostics, employing radionuclides for both imaging and therapy, with prostate-specific membrane antigen (PSMA) at the forefront. Clinical milestones have been reached, including FDA- and/or EMA-approved PSMA-targeted radiodiagnostic agents, such as [18F]DCFPyL (PYLARIFY®, Lantheus Holdings), [18F]rhPSMA-7.3 (POSLUMA®, Blue Earth Diagnostics) and [68Ga]Ga-PSMA-11 (Locametz®, Novartis/ ILLUCCIX®, Telix Pharmaceuticals), as well as PSMA-targeted radiotherapeutic agents, such as [177Lu]Lu-PSMA-617 (Pluvicto®, Novartis). Concurrently, ligand-drug and immune therapies designed to target PSMA are being advanced through rigorous preclinical research and clinical trials. This review delves into the annals of PSMA-targeted radiotheranostics, exploring its historical evolution as a signature molecule in PCa management. We scrutinise its clinical ramifications, acknowledge its limitations, and peer into the avenues that need further exploration. In the crucible of scientific inquiry, we aim to illuminate the path toward a future where the enigma of PCa is deciphered and where its menace is met with precise and effective countermeasures. In the following sections, we discuss the intriguing terrain of PCa radiotheranostics through the lens of PSMA, with the fervent hope of advancing our understanding and enhancing clinical practice.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Prostatic Neoplasms , Radiopharmaceuticals , Humans , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/therapy , Glutamate Carboxypeptidase II/metabolism , Male , Antigens, Surface/metabolism , Radiopharmaceuticals/therapeutic use , Nuclear Medicine/methods , Nuclear Medicine/history , Theranostic Nanomedicine/methods , Radioisotopes/therapeutic use , History, 21st Century , History, 20th Century
10.
Int J Nanomedicine ; 19: 4995-5010, 2024.
Article in English | MEDLINE | ID: mdl-38832336

ABSTRACT

Introduction: Prostate cancer (PC) is the second most common cancer and the fifth most frequent cause of cancer death among men. Prostate-specific membrane antigen (PSMA) expression is associated with aggressive PC, with expression in over 90% of patients with metastatic disease. Those characteristics have led to its use for PC diagnosis and therapies with radiopharmaceuticals, antibody-drug conjugates, and nanoparticles. Despite these advancements, none of the current therapeutics are curative and show some degree of toxicity. Here we present the synthesis and preclinical evaluation of a multimodal, PSMA-targeted dendrimer-drug conjugate (PT-DDC), synthesized using poly(amidoamine) (PAMAM) dendrimers. PT-DDC was designed to enable imaging of drug delivery, providing valuable insights to understand and enhance therapeutic response. Methods: The PT-DDC was synthesized through consecutive conjugation of generation-4 PAMAM dendrimers with maytansinoid-1 (DM1) a highly potent antimitotic agent, Cy5 infrared dye for optical imaging, 2,2',2"-(1,4,7-triazacyclononane-1,4,7-triyl)triacetic acid (NOTA) chelator for radiolabeling with copper-64 and positron emission tomography tomography/computed tomography (PET/CT), lysine-urea-glutamate (KEU) PSMA-targeting moiety and the remaining terminal primary amines were capped with butane-1,2-diol. Non-targeted control dendrimer-drug conjugate (Ctrl-DDC) was formulated without conjugation of KEU. PT-DDC and Ctrl-DDC were characterized using high-performance liquid chromatography, matrix assisted laser desorption ionization mass spectrometry and dynamic light scattering. In vitro and in vivo evaluation of PT-DDC and Ctrl-DDC were carried out in isogenic human prostate cancer PSMA+ PC3 PIP and PSMA- PC3 flu cell lines, and in mice bearing the corresponding xenografts. Results: PT-DDC was stable in 1×PBS and human blood plasma and required glutathione for DM1 release. Optical, PET/CT and biodistribution studies confirmed the in vivo PSMA-specificity of PT-DDC. PT-DDC demonstrated dose-dependent accumulation and cytotoxicity in PSMA+ PC3 PIP cells, and also showed growth inhibition of the corresponding tumors. PT-DDC did not accumulate in PSMA- PC3 flu tumors and did not inhibit their growth. Ctrl-DDC did not show PSMA specificity. Conclusion: In this study, we synthesized a multimodal theranostic agent capable of delivering DM1 and a radionuclide to PSMA+ tumors. This approach holds promise for enhancing image-guided treatment of aggressive, metastatic subtypes of prostate cancer.


Subject(s)
Antigens, Surface , Dendrimers , Glutamate Carboxypeptidase II , Prostatic Neoplasms , Dendrimers/chemistry , Dendrimers/pharmacokinetics , Dendrimers/pharmacology , Male , Humans , Glutamate Carboxypeptidase II/metabolism , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Antigens, Surface/metabolism , Cell Line, Tumor , Animals , Mice , Positron Emission Tomography Computed Tomography/methods , Drug Delivery Systems/methods
11.
Bioorg Med Chem ; 108: 117773, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38850999

ABSTRACT

In this study, we have developedsmall molecule drug conjugates (SMDCs)consisting ofa prostate specific membrane antigen (PSMA) ligandand syringolin derivatives, which are potent proteasome inhibitors, to selectively deliver syringolin derivatives to prostate cancer cells. Two parent compounds were used for syringolin derivatives with different linkage sites. These SMDCs exhibited PSMA-expressing cell-selective cytotoxicity and they could potentially be used for safer treatment of cancer.


Subject(s)
Antigens, Surface , Antineoplastic Agents , Glutamate Carboxypeptidase II , Proteasome Inhibitors , Humans , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/chemical synthesis , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antigens, Surface/metabolism , Structure-Activity Relationship , Molecular Structure , Drug Screening Assays, Antitumor , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism
12.
Eur J Med Chem ; 274: 116545, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38823263

ABSTRACT

Prostate cancer (PCa) is one of the most common tumors in men, with the overexpression of prostate-specific membrane. In this study, we developed four new 68Ga-labeled PSMA-targeting tracers by introducing quinoline, phenylalanine and decanoic acid groups to enhance their lipophilicity, strategically limiting their metabolic pathway through the urinary system. Four radiotracers were synthesized with radiochemical purity >95 %, and exhibited high stability in vivo and in vitro. The inhibition constants (Ki) of SDTWS01-04 to PSMA were in the nanomolar range (<10 nM). Micro PET/CT imaging and biodistribution analysis revealed that 68Ga-SDTWS01 enabled clear tumor visualization in PET images at 1.5 h post-injection, with excellent pharmacokinetic properties. Notably, the kidney uptake of 68Ga-SDTWS01 significantly reduced, with higher tumor-to-kidney ratio (0.36 ± 0.02), tumor-to-muscle ratio (24.31 ± 2.10), compared with 68Ga-PSMA-11 (T/K: 0.15 ± 0.01; T/M: 14.97 ± 1.40), suggesting that 68Ga-SDTWS01 is a promising radiotracer for the diagnosis of PCa. Moreover, SDTWS01 with a chelator DOTA could also label 177Lu and 225Ac, which could be used for the treatment of PCa.


Subject(s)
Gallium Radioisotopes , Glutamate Carboxypeptidase II , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms , Gallium Radioisotopes/chemistry , Humans , Male , Animals , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Glutamate Carboxypeptidase II/metabolism , Glutamate Carboxypeptidase II/antagonists & inhibitors , Tissue Distribution , Mice , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Antigens, Surface/metabolism , Molecular Structure , Cell Line, Tumor
13.
Nucl Med Commun ; 45(7): 622-628, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38835182

ABSTRACT

AIM: The early detection of prostate cancer (PCa) metastatic disease with PET imaging leads to stage migration and change of disease management. We aimed to assess the impact on clinical management deriving from prostate-specific membrane antigen (PSMA) imaging with a digital PET/CT during the routine application in the staging and restaging process of PCa. MATERIAL AND METHODS: Eighty consecutive PCa patients underwent 18F-PSMA-1007. Digital PET/CT were retrospectively evaluated and discussed with oncologists to evaluate the impact on clinical management. Performances analysis, correlation among variables also considering semiquantitative parameters have been conducted. RESULTS: In the whole group of 80 patients at staging (N = 31) and restaging (N = 49), the detection rate of PSMA PET was 85% for all lesions. At staging, the performance analysis resulted in sensitivity 77.6%, specificity 89.5%, negative predictive value (NPV) 77.6%, positive predictive value (PPV) 89.5%, accuracy 85.7%, and area under curve (AUC) 0.87%. The performance of restaging PET in the group of patients with PSA values <1 ng/ml resulted in the following values: sensitivity 66.7%, specificity 92.9%, NPV 85.7%, PPV 81.3%, accuracy 82.6%, and AUC 0.79. Semiquantitative analysis revealed a mean value of SUVmax, metabolic tumor volume, and total lesion PSMA expression with differences in patients with high risk compared to low intermediate. At restaging PET, semiquantitative values of patients with total prostate specific antigen (tPSA) ≤ 1 ng/ml were significantly less than those of the tPSA > 1 ng/ml. A significant impact on clinical management was reported in 46/80 patients (57.5%) based on PSMA PET findings at staging and restaging. CONCLUSION: Although PSMA-PET provides optimal performances, its current role in redefining a better staging should be translated in the current clinical scenario about potential improvement in clinical/survival outcomes.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Neoplasm Staging , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Aged , Middle Aged , Retrospective Studies , Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Aged, 80 and over , Oligopeptides , Niacinamide/analogs & derivatives
14.
Animal Model Exp Med ; 7(3): 337-346, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38859745

ABSTRACT

BACKGROUND: This study aimed to construct and characterize a humanized influenza mouse model expressing hST6GAL1. METHODS: Humanized fragments, consisting of the endothelial cell-specific K18 promoter, human ST6GAL1-encoding gene, and luciferase gene, were microinjected into the fertilized eggs of mice. The manipulated embryos were transferred into the oviducts of pseudopregnant female mice. The offspring were identified using PCR. Mice exhibiting elevated expression of the hST6GAL1 gene were selectively bred for propagation, and in vivo analysis was performed for screening. Expression of the humanized gene was tested by performing immunohistochemical (IHC) analysis. Hematologic and biochemical analyses using the whole blood and serum of humanized hST6GAL1 mice were performed. RESULTS: Successful integration of the human ST6GAL1 gene into the mouse genome led to the overexpression of human SiaT ST6GAL1. Seven mice were identified as carrying copies of the humanized gene, and the in vivo analysis indicated that hST6GAL1 gene expression in positive mice mirrored influenza virus infection characteristics. The IHC results revealed that hST6GAL1 was expressed in the lungs of humanized mice. Moreover, the hematologic and biochemical parameters of the positive mice were within the normal range. CONCLUSION: A humanized influenza mouse model expressing the hST6GAL1 gene was successfully established and characterized.


Subject(s)
Disease Models, Animal , Sialyltransferases , Animals , Humans , Mice , Female , Sialyltransferases/genetics , Sialyltransferases/metabolism , Antigens, CD/metabolism , Antigens, CD/genetics , Orthomyxoviridae Infections , Mice, Transgenic , Antigens, Surface/metabolism , Antigens, Surface/genetics , beta-D-Galactoside alpha 2-6-Sialyltransferase
15.
Theranostics ; 14(7): 2736-2756, 2024.
Article in English | MEDLINE | ID: mdl-38773975

ABSTRACT

Radical prostatectomy (RP) combined with pelvic lymph node dissection (PLND) is the first step in multimodal treatment of prostate cancer (PCa) without distant metastases. For a long time, the surgical resection range has been highly dependent on the surgeon's visualization and experience with preoperative imaging. With the rapid development of prostate-specific membrane antigen positron emission tomography and single-photon emission computed tomography (PSMA-PET and PSMA-SPECT), PSMA-targeted surgery has been introduced for a more accurate pathological diagnosis and complete resection of positive surgical margins (PSMs) and micro-lymph node metastases (LNMs). We reviewed PSMA-targeted surgeries, including PSMA-PET-guided prostatic biopsy (PSMA-TB), PSMA-targeted radio-guided surgery (PSMA-RGS), PSMA-targeted fluorescence-guided surgery (PSMA-FGS), and multi-modality/multi-targeted PSMA-targeted surgery. We also discuss the strengths and challenges of PSMA-targeted surgery, and propose that PSMA-targeted surgery could be a great addition to existing surgery protocols, thereby improving the accuracy and convenience of surgery for primary and recurrent PCa in the near future.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Prostatectomy , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/surgery , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Prostatectomy/methods , Surgery, Computer-Assisted/methods , Positron-Emission Tomography/methods , Tomography, Emission-Computed, Single-Photon/methods , Lymph Node Excision/methods
16.
Nature ; 630(8015): 206-213, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778111

ABSTRACT

Targeted radionuclide therapy, in which radiopharmaceuticals deliver potent radionuclides to tumours for localized irradiation, has addressed unmet clinical needs and improved outcomes for patients with cancer1-4. A therapeutic radiopharmaceutical must achieve both sustainable tumour targeting and fast clearance from healthy tissue, which remains a major challenge5,6. A targeted ligation strategy that selectively fixes the radiopharmaceutical to the target protein in the tumour would be an ideal solution. Here we installed a sulfur (VI) fluoride exchange (SuFEx) chemistry-based linker on radiopharmaceuticals to prevent excessively fast tumour clearance. When the engineered radiopharmaceutical binds to the tumour-specific protein, the system undergoes a binding-to-ligation transition and readily conjugates to the tyrosine residues through the 'click' SuFEx reaction. The application of this strategy to a fibroblast activation protein (FAP) inhibitor (FAPI) triggered more than 80% covalent binding to the protein and almost no dissociation for six days. In mice, SuFEx-engineered FAPI showed 257% greater tumour uptake than did the original FAPI, and increased tumour retention by 13-fold. The uptake in healthy tissues was rapidly cleared. In a pilot imaging study, this strategy identified more tumour lesions in patients with cancer than did other methods. SuFEx-engineered FAPI also successfully achieved targeted ß- and α-radionuclide therapy, causing nearly complete tumour regression in mice. Another SuFEx-engineered radioligand that targets prostate-specific membrane antigen (PSMA) also showed enhanced therapeutic efficacy. Considering the broad scope of proteins that can potentially be ligated to SuFEx warheads, it might be possible to adapt this strategy to other cancer targets.


Subject(s)
Molecular Targeted Therapy , Prostatic Neoplasms , Radioisotopes , Radiopharmaceuticals , Animals , Humans , Male , Mice , Antigens, Surface/chemistry , Antigens, Surface/metabolism , Cell Line, Tumor , Fluorides/chemistry , Fluorides/metabolism , Glutamate Carboxypeptidase II/chemistry , Glutamate Carboxypeptidase II/metabolism , Ligands , Membrane Proteins/metabolism , Membrane Proteins/chemistry , Molecular Targeted Therapy/methods , Pilot Projects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Sulfur Compounds/chemistry , Sulfur Compounds/metabolism , Tyrosine/metabolism , Tyrosine/chemistry , Xenograft Model Antitumor Assays
17.
J Nucl Med ; 65(7): 1007-1012, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38782453

ABSTRACT

Prostate-specific membrane antigen (PSMA) is expressed in the neovasculature of multiple solid tumors, including renal cell carcinoma (RCC). Studies have demonstrated promising results on the utility of PSMA-targeted PET/CT imaging in RCC. This report aims to provide a systematic review and metaanalysis on the utility and detection rate of PSMA PET/CT imaging in staging or evaluation of primary RCC and restaging of metastatic or recurrent RCC. Methods: Searches were performed in PubMed, Embase, and abstract proceedings (last updated, August 2023). Studies that provided a lesion-level detection rate of PSMA radiotracers in staging or restaging of RCC were included in the metaanalysis. The overall pooled detection rate with a 95% CI was estimated, and subgroup analysis was performed when feasible. Results: Nine studies comprising 152 patients (133 clear cell RCC [ccRCC], 19 other RCC subtypes) were included in the metaanalysis. The pooled detection rate of PSMA PET/CT in evaluation of primary or metastatic RCC was estimated to be 0.83 (95% CI, 0.67-0.92). Subgroup analysis showed a pooled PSMA detection rate of 0.74 (95% CI, 0.57-0.86) in staging or evaluation of primary RCC lesions and 0.87 (95% CI, 0.73-0.95) in restaging of metastatic or recurrent RCC. Analysis based on the type of radiotracer showed a pooled detection rate of 0.85 (95% CI, 0.62-0.95) for 68Ga-based PSMA tracers and 0.92 (95% CI, 0.76-0.97) for 18F-DCFPyL PET/CT. Furthermore, in metastatic ccRCC, the available data support a significantly higher detection rate for 18F-DCFPyL PET/CT than for conventional imaging modalities (2 studies). Conclusion: Our preliminary results show that PSMA PET/CT could be a promising alternative imaging modality for evaluating RCC, particularly metastatic ccRCC. Large prospective studies are warranted to confirm clinical utility in the staging and restaging of RCC.


Subject(s)
Antigens, Surface , Carcinoma, Renal Cell , Glutamate Carboxypeptidase II , Kidney Neoplasms , Neoplasm Staging , Positron Emission Tomography Computed Tomography , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/pathology , Positron Emission Tomography Computed Tomography/methods , Humans , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/pathology , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism
18.
Radiol Med ; 129(6): 901-911, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38700556

ABSTRACT

PURPOSE: High PSMA expression might be correlated with structural characteristics such as growth patterns on histopathology, not recognized by the human eye on MRI images. Deep structural image analysis might be able to detect such differences and therefore predict if a lesion would be PSMA positive. Therefore, we aimed to train a neural network based on PSMA PET/MRI scans to predict increased prostatic PSMA uptake based on the axial T2-weighted sequence alone. MATERIAL AND METHODS: All patients undergoing simultaneous PSMA PET/MRI for PCa staging or biopsy guidance between April 2016 and December 2020 at our institution were selected. To increase the specificity of our model, the prostatic beds on PSMA PET scans were dichotomized in positive and negative regions using an SUV threshold greater than 4 to generate a PSMA PET map. Then, a C-ENet was trained on the T2 images of the training cohort to generate a predictive prostatic PSMA PET map. RESULTS: One hundred and fifty-four PSMA PET/MRI scans were available (133 [68Ga]Ga-PSMA-11 and 21 [18F]PSMA-1007). Significant cancer was present in 127 of them. The whole dataset was divided into a training cohort (n = 124) and a test cohort (n = 30). The C-ENet was able to predict the PSMA PET map with a dice similarity coefficient of 69.5 ± 15.6%. CONCLUSION: Increased prostatic PSMA uptake on PET might be estimated based on T2 MRI alone. Further investigation with larger cohorts and external validation is needed to assess whether PSMA uptake can be predicted accurately enough to help in the interpretation of mpMRI.


Subject(s)
Deep Learning , Magnetic Resonance Imaging , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Aged , Magnetic Resonance Imaging/methods , Middle Aged , Prostate/diagnostic imaging , Positron-Emission Tomography/methods , Retrospective Studies , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Predictive Value of Tests , Organ Size , Gallium Radioisotopes , Radiopharmaceuticals/pharmacokinetics
19.
Anticancer Res ; 44(6): 2297-2305, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821587

ABSTRACT

BACKGROUND/AIM: The current systematic review aimed to collect and analyze all available published and unpublished cases in which prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (177Lu-PSMA) was used to treat non-prostatic cancer. MATERIALS AND METHODS: Literature search and evidence acquisition through contacts with organizations that use 177Lu-PSMA were employed. PubMed/Medline, SCOPUS, and ScienceDirect searches were performed following PRISMA recommendations. The search strategy was to screen all articles describing 177Lu-PSMA radioligand therapy published to date with the key word "177Lu-PSMA". These articles were collected and screened for non-prostatic cancer cases. Quality assessment was performed using the GRADE criteria. RESULTS: A total of 713 articles were screened, and the search revealed 15 eligible records. Forty patients with a mean age of 51.2±18.5 years were treated with 177Lu-PSMA for non-prostatic cancer. Of them, 30 cases were published, and 10 were found in medical institution records. Cancers of the salivary glands were most often targeted (13/40), followed by various brain cancer types (8/40), and osteosarcoma (6/40). The authors used previously established protocols for castration-resistant prostate cancer with the dose per cycle as 6.0-7.4 GBq and the number of cycles between one and four. Toxicity was estimated as low, and 21 out of 28 patients with reported outcomes survived to the time of the publication. CONCLUSION: PSMA-targeted radioligand therapy was infrequently used to treat different non-prostatic cancer types in various target organs. These pioneering efforts indicate that 177Lu-PSMA can be used to treat non-prostatic cancer with PSMA expression. The toxicity of such treatment was low, and the outcome was relatively good.


Subject(s)
Lutetium , Humans , Lutetium/therapeutic use , Middle Aged , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/adverse effects , Male , Neoplasms/radiotherapy , Neoplasms/therapy , Dipeptides/therapeutic use , Female , Glutamate Carboxypeptidase II/metabolism , Aged , Radioisotopes/therapeutic use , Radioisotopes/adverse effects , Antigens, Surface/metabolism , Adult , Heterocyclic Compounds, 1-Ring/therapeutic use , Prostate-Specific Antigen
20.
Clin Nucl Med ; 49(7): 652-654, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38689438

ABSTRACT

ABSTRACT: Dedifferentiated liposarcoma is an extremely rare and highly malignant tumor. We demonstrated a case of a 75-year-old man with significantly PSMA-avid and mildly FDG uptake-dedifferentiated liposarcoma in the retroperitoneal area. The double-tracer (PSMA and FDG) PET scans could further contribute to differential diagnosis and the following treatment strategy for patients who were suspected with prostate cancer metastases and other malignant tumors simultaneously.


Subject(s)
Fluorodeoxyglucose F18 , Liposarcoma , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms , Humans , Male , Liposarcoma/diagnostic imaging , Liposarcoma/secondary , Aged , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Multimodal Imaging , Neoplasm Metastasis , Tomography, X-Ray Computed , Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...