Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Commun Biol ; 5(1): 1260, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396952

RESUMO

Astatine-211-parthanatine ([211At]PTT) is an alpha-emitting radiopharmaceutical therapeutic that targets poly(adenosine-diphosphate-ribose) polymerase 1 (PARP1) in cancer cells. High-risk neuroblastomas exhibit among the highest PARP1 expression across solid tumors. In this study, we evaluated the efficacy of [211At]PTT using 11 patient-derived xenograft (PDX) mouse models of high-risk neuroblastoma, and assessed hematological and marrow toxicity in a CB57/BL6 healthy mouse model. We observed broad efficacy in PDX models treated with [211At]PTT at the maximum tolerated dose (MTD 36 MBq/kg/fraction x4) administered as a fractionated regimen. For the MTD, complete tumor response was observed in 81.8% (18 of 22) of tumors and the median event free survival was 72 days with 30% (6/20) of mice showing no measurable tumor >95 days. Reversible hematological and marrow toxicity was observed 72 hours post-treatment at the MTD, however full recovery was evident by 4 weeks post-therapy. These data support clinical development of [211At]PTT for high-risk neuroblastoma.


Assuntos
Neuroblastoma , Humanos , Animais , Camundongos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Modelos Animais de Doenças
2.
Clin Cancer Res ; 28(18): 4146-4157, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35861867

RESUMO

PURPOSE: [131I]meta-iodobenzylguanidine ([131I]MIBG) is a targeted radiotherapeutic administered systemically to deliver beta particle radiation in neuroblastoma. However, relapses in the bone marrow are common. [211At]meta-astatobenzylguanidine ([211At] MABG) is an alpha particle emitter with higher biological effectiveness and short path length which effectively sterilizes microscopic residual disease. Here we investigated the safety and antitumor activity [211At]MABG in preclinical models of neuroblastoma. EXPERIMENTAL DESIGN: We defined the maximum tolerated dose (MTD), biodistribution, and toxicity of [211At]MABG in immunodeficient mice in comparison with [131I]MIBG. We compared the antitumor efficacy of [211At]MABG with [131I]MIBG in three murine xenograft models. Finally, we explored the efficacy of [211At]MABG after tail vein xenografting designed to model disseminated neuroblastoma. RESULTS: The MTD of [211At]MABG was 66.7 MBq/kg (1.8 mCi/kg) in CB17SC scid-/- mice and 51.8 MBq/kg (1.4 mCi/kg) in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. Biodistribution of [211At]MABG was similar to [131I]MIBG. Long-term toxicity studies on mice administered with doses up to 41.5 MBq/kg (1.12 mCi/kg) showed the radiotherapeutic to be well tolerated. Both 66.7 MBq/kg (1.8 mCi/kg) single dose and fractionated dosing 16.6 MBq/kg/fraction (0.45 mCi/kg) × 4 over 11 days induced marked tumor regression in two of the three models studied. Survival was significantly prolonged for mice treated with 12.9 MBq/kg/fraction (0.35 mCi/kg) × 4 doses over 11 days [211At]MABG in the disseminated disease (IMR-05NET/GFP/LUC) model (P = 0.003) suggesting eradication of microscopic disease. CONCLUSIONS: [211At]MABG has significant survival advantage in disseminated models of neuroblastoma. An alpha particle emitting radiopharmaceutical may be effective against microscopic disseminated disease, warranting clinical development.


Assuntos
Astato , Neuroblastoma , 3-Iodobenzilguanidina/efeitos adversos , Partículas alfa/uso terapêutico , Animais , Astato/uso terapêutico , Guanidinas/uso terapêutico , Humanos , Radioisótopos do Iodo/uso terapêutico , Camundongos , Camundongos Endogâmicos NOD , Recidiva Local de Neoplasia/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Neuroblastoma/radioterapia , Compostos Radiofarmacêuticos/efeitos adversos , Distribuição Tecidual , Células Tumorais Cultivadas
3.
Mol Ther ; 28(1): 42-51, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31668558

RESUMO

Cell-based therapeutics have considerable promise across diverse medical specialties; however, reliable human imaging of the distribution and trafficking of genetically engineered cells remains a challenge. We developed positron emission tomography (PET) probes based on the small-molecule antibiotic trimethoprim (TMP) that can be used to image the expression of the Escherichia coli dihydrofolate reductase enzyme (eDHFR) and tested the ability of [18F]-TMP, a fluorine-18 probe, to image primary human chimeric antigen receptor (CAR) T cells expressing the PET reporter gene eDHFR, yellow fluorescent protein (YFP), and Renilla luciferase (rLuc). Engineered T cells showed an approximately 50-fold increased bioluminescent imaging signal and 10-fold increased [18F]-TMP uptake compared to controls in vitro. eDHFR-expressing anti-GD2 CAR T cells were then injected into mice bearing control GD2- and GD2+ tumors. PET/computed tomography (CT) images acquired on days 7 and 13 demonstrated early residency of CAR T cells in the spleen followed by on-target redistribution to the GD2+ tumors. This was corroborated by autoradiography and anti-human CD8 immunohistochemistry. We found a high sensitivity of detection for identifying tumor-infiltrating CD8 CAR T cells, ∼11,000 cells per mm3. These data suggest that the [18F]-TMP/eDHFR PET pair offers important advantages that could better allow investigators to monitor immune cell trafficking to tumors in patients.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Escherichia coli/enzimologia , Genes Reporter , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Receptores de Antígenos Quiméricos/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Animais , Linfócitos T CD8-Positivos/metabolismo , Feminino , Radioisótopos de Flúor , Gangliosídeos/metabolismo , Células HCT116 , Voluntários Saudáveis , Xenoenxertos/diagnóstico por imagem , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Baço/diagnóstico por imagem , Baço/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima
4.
Mol Cancer Ther ; 18(7): 1195-1204, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31072830

RESUMO

Alpha-emitters can be pharmacologically delivered for irradiation of single cancer cells, but cellular lethality could be further enhanced by targeting alpha-emitters directly to the nucleus. PARP-1 is a druggable protein in the nucleus that is overexpressed in neuroblastoma compared with normal tissues and is associated with decreased survival in high-risk patients. To exploit this, we have functionalized a PARP inhibitor (PARPi) with an alpha-emitter astatine-211. This approach offers enhanced cytotoxicity from conventional PARPis by not requiring enzymatic inhibition of PARP-1 to elicit DNA damage; instead, the alpha-particle directly induces multiple double-strand DNA breaks across the particle track. Here, we explored the efficacy of [211At]MM4 in multiple cancers and found neuroblastoma to be highly sensitive in vitro and in vivo Furthermore, alpha-particles delivered to neuroblastoma show antitumor effects and durable responses in a neuroblastoma xenograft model, especially when administered in a fractionated regimen. This work provides the preclinical proof of concept for an alpha-emitting drug conjugate that directly targets cancer chromatin as a therapeutic approach for neuroblastoma and perhaps other cancers.


Assuntos
Neuroblastoma/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Análise de Sobrevida
5.
Clin Cancer Res ; 25(10): 3063-3073, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30692100

RESUMO

PURPOSE: Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors in combination with endocrine-therapy have emerged as an important regimen of care for estrogen receptor (ER)-positive metastatic breast cancer, although identifying predictive biomarkers remains a challenge. We assessed the ability of two PET-proliferation tracers, [18F]FLT and [18F]ISO-1, for evaluating response to CDK4/6-inhibitor (palbociclib) and ER-antagonist (fulvestrant). EXPERIMENTAL DESIGN: To determine the effect of CDK4/6 inhibition combined with estrogen-blockade, we assessed cell proliferation in six breast cancer cell lines after 1, 3, and 6 days of treatment with palbociclib and/or fulvestrant. These data were correlated to in vitro radiotracer assays and results were verified by longitudinal [18F]FLT and [18F]ISO-1 micro-PET imaging performed in MCF7 tumor-bearing mice. RESULTS: All palbociclib-sensitive cell lines showed decreased [18F]FLT accumulation and S-phase depletion after treatment, with both measures augmented by combination therapy. In contrast, these cells showed changes in [18F]ISO-1 analogue-binding and G0 arrest only after prolonged treatment. MicroPET imaging of MCF7 xenografts showed a significant decrease in [18F]FLT but no changes in [18F]ISO-1 uptake in all treated mice on day 3. On day 14, however, mice treated with combination therapy showed a significant decrease in [18F]ISO-1, corresponding to G0 arrest, while maintaining reduced [18F]FLT uptake, which corresponded to S-phase depletion. CONCLUSIONS: Our data suggest complementary roles of [18F]FLT and [18F]ISO-1 PET in evaluating tumor-proliferation after combined CDK4/6 inhibitor and endocrine therapy in breast cancer. [18F]FLT is more sensitive to immediate changes in S-phase, whereas [18F]ISO-1 can assess more delayed changes related to cell-cycle arrest and transition to G0 quiescence from combination therapy. These data suggest a potential role for early prediction of long-term response using these imaging biomarkers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Didesoxinucleosídeos , Antagonistas do Receptor de Estrogênio/administração & dosagem , Feminino , Radioisótopos de Flúor , Fulvestranto/administração & dosagem , Humanos , Estudos Longitudinais , Células MCF-7 , Camundongos , Camundongos SCID , Piperazinas/administração & dosagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Piridinas/administração & dosagem , Compostos Radiofarmacêuticos , Distribuição Aleatória , Receptores de Estrogênio/antagonistas & inibidores , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Bioorg Chem ; 83: 242-249, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30390553

RESUMO

Poly(ADP-ribose)polymerase-1 inhibitor (PARPi) AZD2461 was designed to be a weak P-glycoprotein (P-gp) analogue of FDA approved olaparib. With this chemical property in mind, we utilized the AZD2461 ligand architecture to develop a CNS penetrant and PARP-1 selective imaging probe, in order to investigate PARP-1 mediated neuroinflammation and neurodegenerative diseases, such as Alzheimer's and Parkinson's. Our work led to the identification of several high-affinity PARPi, including AZD2461 congener 9e (PARP-1 IC50 = 3.9 ±â€¯1.2 nM), which was further evaluated as a potential 18F-PET brain imaging probe. However, despite the similar molecular scaffolds of 9e and AZD2461, our studies revealed non-appreciable brain-uptake of [18F]9e in non-human primates, suggesting AZD2461 to be non-CNS penetrant.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Ftalazinas/farmacologia , Piperidinas/farmacologia , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/agonistas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Radioisótopos de Flúor/química , Humanos , Macaca mulatta , Masculino , Camundongos Endogâmicos BALB C , Ftalazinas/síntese química , Piperidinas/síntese química
7.
EJNMMI Phys ; 5(1): 21, 2018 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-30294746

RESUMO

BACKGROUND: [18F]Fluortriopride (FTP) was developed as a dopamine D3-selective radiotracer, thought to be important to neurobiological reward pathways and implicated in drug addiction, Parkinson's disease, and schizophrenia. Preclinical radiation dosimetry studies found the gallbladder wall received the highest dose. A gallbladder dose reduction intervention was simulated using a novel reduction model for healthy adults following fatty-meal consumption. The goals of this study were to assess whole body FTP human dosimetry and determine the feasibility of reducing absorbed dose to the gallbladder wall. RESULTS: Effective dose without a fatty meal was 0.022 ± 0.002 mSv/MBq (± standard deviation) with highest organ dose of 0.436 ± 0.178 mSv/MBq to the gallbladder wall (n = 10). Predicted gallbladder dose reduction with fatty meal consumed was 67.4% (n = 10). Meal consumption by four repeat volunteers decreased average gallbladder dose by 71.3% (n = 4) compared to the original ten volunteers. CONCLUSIONS: Observed effective doses were adequately low to continue studying FTP uptake in humans. Validated dosimetry simulations indicate up to a 71% reduction in gallbladder dose can be achieved by employing intrinsic physiology to contract the gallbladder via fatty meal ingestion. This methodology for predicting gallbladder absorbed dose reduction from fatty meal consumption can be applied to other radiopharmaceuticals and radiotherapies.

8.
J Clin Invest ; 128(5): 2116-2126, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29509546

RESUMO

BACKGROUND: Poly(ADP-ribose) polymerase (PARP) inhibitors are effective in a broad population of patients with ovarian cancer; however, resistance caused by low enzyme expression of the drug target PARP-1 remains to be clinically evaluated in this context. We hypothesize that PARP-1 expression is variable in ovarian cancer and can be quantified in primary and metastatic disease using a novel PET imaging agent. METHODS: We used a translational approach to describe the significance of PET imaging of PARP-1 in ovarian cancer. First, we produced PARP1-KO ovarian cancer cell lines using CRISPR/Cas9 gene editing to test the loss of PARP-1 as a resistance mechanism to all clinically used PARP inhibitors. Next, we performed preclinical microPET imaging studies using ovarian cancer patient-derived xenografts in mouse models. Finally, in a phase I PET imaging clinical trial we explored PET imaging as a regional marker of PARP-1 expression in primary and metastatic disease through correlative tissue histology. RESULTS: We found that deletion of PARP1 causes resistance to all PARP inhibitors in vitro, and microPET imaging provides proof of concept as an approach to quantify PARP-1 in vivo. Clinically, we observed a spectrum of standard uptake values (SUVs) ranging from 2-12 for PARP-1 in tumors. In addition, we found a positive correlation between PET SUVs and fluorescent immunohistochemistry for PARP-1 (r2 = 0.60). CONCLUSION: This work confirms the translational potential of a PARP-1 PET imaging agent and supports future clinical trials to test PARP-1 expression as a method to stratify patients for PARP inhibitor therapy. TRIAL REGISTRATION: Clinicaltrials.gov NCT02637934. FUNDING: Research reported in this publication was supported by the Department of Defense OC160269, a Basser Center team science grant, NIH National Cancer Institute R01CA174904, a Department of Energy training grant DE-SC0012476, Abramson Cancer Center Radiation Oncology pilot grants, the Marsha Rivkin Foundation, Kaleidoscope of Hope Foundation, and Paul Calabresi K12 Career Development Award 5K12CA076931.


Assuntos
Meios de Contraste/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Neoplasias Ovarianas , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Camundongos , Pessoa de Meia-Idade , Proteínas de Neoplasias , Transplante de Neoplasias , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo
9.
Proc Natl Acad Sci U S A ; 114(31): 8372-8377, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28716936

RESUMO

There is often overlap in the diagnostic features of common pathologic processes such as infection, sterile inflammation, and cancer both clinically and using conventional imaging techniques. Here, we report the development of a positron emission tomography probe for live bacterial infection based on the small-molecule antibiotic trimethoprim (TMP). [18F]fluoropropyl-trimethoprim, or [18F]FPTMP, shows a greater than 100-fold increased uptake in vitro in live bacteria (Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa) relative to controls. In a rodent myositis model, [18F]FPTMP identified live bacterial infection without demonstrating confounding increased signal in the same animal from other etiologies including chemical inflammation (turpentine) and cancer (breast carcinoma). Additionally, the biodistribution of [18F]FPTMP in a nonhuman primate shows low background in many important tissues that may be sites of infection such as the lungs and soft tissues. These results suggest that [18F]FPTMP could be a broadly useful agent for the sensitive and specific imaging of bacterial infection with strong translational potential.


Assuntos
Antibacterianos/farmacologia , Infecções por Escherichia coli/diagnóstico , Escherichia coli/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Infecções por Pseudomonas/diagnóstico , Pseudomonas aeruginosa/metabolismo , Infecções Estafilocócicas/diagnóstico , Staphylococcus aureus/metabolismo , Trimetoprima/farmacologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Radioisótopos de Flúor/química , Células HCT116 , Humanos , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Infecções por Pseudomonas/microbiologia , Compostos Radiofarmacêuticos/farmacologia , Infecções Estafilocócicas/microbiologia , Trimetoprima/química
10.
Cancer Biother Radiopharm ; 32(1): 9-15, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28118040

RESUMO

INTRODUCTION: Poly (ADP-ribose) polymerase 1 (PARP-1) is the major target of clinical PARP inhibitors and is a potential predictive biomarker for response to therapy. Due to the limited success of PARP inhibitors as monotherapy, investigators have shifted the clinical role of PARP inhibitors to the adjuvant setting. In this study, we evaluate the radiotracer [18F]FluorThanatrace ([18F]FTT) as a marker of PARP expression in vitro and the associated biological implications of PARP-1 expression in PARP inhibitor treatment adjuvant to radiation therapy. MATERIALS AND METHODS: SNU-251 (BRCA1-mutant) and SKOV3 (BRCA1-WT) cell lines were evaluated in vitro by using the radiotracer [18F]FTT. Pharmacological binding assays were performed at baseline and were correlated with PARP-1 protein expression measured by Western blot protein analysis. Cell viability and clonogenic assays were used to characterize in vitro cytotoxicity for treatments, including: PARP inhibitors alone, radiation alone, and PARP inhibitor adjuvant to radiation. Western blot protein analysis was used to assess response to treatment by using γH2AX to measure DNA damage and PAR to measure the catalytic inhibition of PARP. RESULTS: [18F]FTT was capable of measuring PARP-1 protein expression in vitro and corresponded to Western blot protein analysis at baseline. The addition of a PARP inhibitor enhanced radiation effects in both cell lines; however, a greater synergy was observed in the SNU-251 cell line that expresses a BRCA1 mutation and homologous recombination deficiency. Western blot protein analysis showed that the addition of a PARP inhibitor adjuvant to radiation increases DNA damage in both cell lines and reduces PARP enzymatic activity as measured by PAR. CONCLUSIONS: In this work, we found that PARP-1 expression positively corresponds in vitro to the response of PARP inhibitors in combination with radiation therapy in ovarian cancer.


Assuntos
Biomarcadores Farmacológicos/metabolismo , Neoplasias Ovarianas/terapia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Quimioterapia Adjuvante , Dano ao DNA/efeitos dos fármacos , Feminino , Radioisótopos de Flúor , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/radioterapia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico
11.
Nucl Med Biol ; 43(11): 721-731, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27589334

RESUMO

INTRODUCTION: Nine novel analogues were synthesized including a 6-carbon spacer analogue of ISO-1 (7). They have moderate binding affinity for sigma-2 (σ2) receptors and high selectivity for σ2 receptors relative to sigma-1 (σ1) receptors. METHODS: ([18F]7) was synthesized and evaluated as a candidate ligand for positron emission (PET) imaging of the σ2 receptor in tumors. Radioligand [18F]7 was radiolabeled with 18F via displacement of the corresponding mesylate precursor with [18F]fluoride. Cellular uptake study of [18F]7 was performed in EMT-6 tumor cell, and in vivo biodistribution study of [18F]7 and microPET imaging study of [18F]3 and [18F]7 carried out in female Balb/c mice bearing EMT-6 tumors. RESULTS: [18F]7 had a respectable tumor uptake (1.55%ID/g at 60min post-injection) and high tumor/muscle ratios at 60 and 120min post-injection. MicroPET imaging of [18F]7 in tumor-bearing mice as above showed significant tumor localization and a high tumor/muscle ratio as well. CONCLUSIONS: These results are similar to or better than [18F]ISO-1 ([18F]3), which indicates that [18F]7 has potential for imaging the σ2 receptor status of solid tumors.


Assuntos
Benzamidas/química , Carbono/química , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Receptores sigma/metabolismo , Tetra-Hidroisoquinolinas/química , Animais , Benzamidas/metabolismo , Benzamidas/farmacocinética , Transporte Biológico , Linhagem Celular Tumoral , Feminino , Camundongos , Conformação Molecular , Radioquímica , Distribuição Tecidual
12.
Am J Nucl Med Mol Imaging ; 6(1): 94-101, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27069769

RESUMO

The nuclear enzyme PARP1 plays a central role in sensing DNA damage and facilitating repair. Tumors with BRCA1/2 mutations are highly dependent on PARP1 as an alternative mechanism for DNA repair, and PARP inhibitors generate synthetic lethality in tumors with BRCA mutations, resulting in cell cycle arrest and apoptosis. Zhou et al. recently synthesized an (18)F-labeled PARP1 inhibitor ([(18)F]FluorThanatrace) for PET, and demonstrated high specific tracer uptake in a xenograft model of breast cancer [1]. In the current study, we characterize the level of baseline PARP expression and activity across multiple human breast cancer cell lines, including a BRCA1 mutant line. PARP expression and activity, as measured by levels of PAR and PARP1, is correlated with in vitro [(18)F]FluorThanatrace binding as well as tracer uptake on PET in a xenograft model of breast cancer. Radiotracer uptake in genetically-engineered mouse fibroblasts indicates [(18)F]FluorThanatrace is selective for PARP1 versus other PARP enzymes. This motivates further studies of [(18)F]FluorThanatrace as an in vivo measure of PARP1 expression and activity in patients who would benefit from PARP inhibitor therapy.

13.
Nucl Med Biol ; 43(1): 35-41, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26702785

RESUMO

RATIONALE: The sigma-2 receptor is a protein with a Heme binding region and is capable of receptor-mediated endocytosis. It is overexpressed in many cancers making it a potential vector for therapeutic drug delivery. Our objective was to introduce an alpha-emitting radionuclide, astatine-211, into a selective sigma-2 ligand moiety to provide cytotoxic capabilities without adversely altering the pharmacological characteristics. In this study we investigated the in vitro/in vivo tumor targeting and estimated dosimetry of alpha-emitting sigma-2 ligand, 5-(astato-(211)At)-N-(4-(6,7-dimethoxy-3,4-dihydroisoquinolin-2(1H)-yl)butyl)-2,3-dimethoxybenzamide ((211)At-MM3), in a pre-clinical human breast cancer model. METHODS: Astatine-211 was produced in a cyclotron and isolated by dry distillation. Radiosynthesis of (211)At-MM3 was performed using a tin precursor through radioastatodestannylation. In vitro sigma-2 binding experiments using (211)At-MM3 were carried out in live EMT6 and MDA-MB-231 breast cancer cells and liver homogenate tissue. In vivo biodistribution experiments were performed using EMT6 mouse breast cancer cells in BALB/c female mice. Approximately 370 kBq of (211)At-MM3 was administered intravenously and at time points of 5 min, 1, 2, 4, 8, and 24 h organs/tissue were harvested. Estimated human dosimetry was extrapolated from biodistribution data using OLINDA/EXM (VU e-Innovations). RESULTS: Astatine-211 was successfully produced and isolated in quantities suitable for in vitro and small animal in vivo experiments. Radiosynthesis of (211)At-MM3 was reproducible with high radiochemical purity. Astatine-211-MM3 exhibited picomolar affinity to the sigma-2 receptor in contrast to the iodinated analog that had nanomolar affinity. Prolonged tumor targeting was measured through biodistribution studies with a maximal tumor to muscle ratio of 9.02 at 4h. Estimated human dosimetry revealed doses of up to 370 MBq in an adult female patient were below organ radiation limits with the potential to provide a high therapeutic dose to tumors. CONCLUSION: The sigma-2 receptor could serve as a suitable targeting platform for designing radiotherapeutics. (211)At-MM3 showed tumor targeting properties in vitro/in vivo and favorable estimated human dosimetry establishing the proof of concept for future development as a radiotherapeutic for the treatment of breast cancer.


Assuntos
Partículas alfa/uso terapêutico , Terapia de Alvo Molecular/métodos , Receptores sigma/metabolismo , Animais , Astato/uso terapêutico , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacocinética , Benzamidas/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Feminino , Humanos , Camundongos , Radiometria , Distribuição Tecidual
14.
Biochem Biophys Res Commun ; 467(4): 1070-5, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26453012

RESUMO

BACKGROUND: Triple-negative breast cancer (TNBC) is associated with high relapse rates and increased mortality when compared with other breast cancer subtypes. In contrast to receptor positive breast cancers, there are no approved targeted therapies for TNBC. Identifying biomarkers for TNBC is of high importance for the advancement of patient care. The sigma-2 receptor has been shown to be overexpressed in triple negative breast cancer in vivo and has been characterized as a marker of proliferation. The aim of the present study was to define the sigma-2 receptor as a target for therapeutic drug delivery and biomarker in TNBC. METHODS: Three TNBC cell lines were evaluated: MDA-MB-231, HCC1937 and HCC1806. Sigma-2 compounds were tested for pharmacological properties specific to the sigma-2 receptor through competitive inhibition assays. Sigma-2 receptor expression was measured through radioligand receptor saturation studies. Drug sensitivity for taxol was compared to a sigma-2 targeting compound conjugated to a cytotoxic payload, SW IV-134. Cell viability was assessed after treatments for 2 or 48 h. Sigma-2 blockade was assessed to define sigma-2 mediated cytotoxicity of SW IV-134. Caspase 3/7 activation induced by SW IV-134 was measured at corresponding treatment time points. RESULTS: SW IV-134 was the most potent compound tested in two of the three cell lines and was similarly effective in all three. MDA-MB-231 displayed a statistically significant higher sigma-2 receptor expression and also was the most sensitive cell line evaluated to SW IV-134. CONCLUSION: Targeting the sigma-2 receptor with a cytotoxic payload was effective in all the three cell lines evaluated and provides the proof of concept for future development of a therapeutic platform for the treatment of TNBC.


Assuntos
Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Receptores sigma/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias de Mama Triplo Negativas/metabolismo
15.
J Nucl Med ; 56(11): 1724-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26272810

RESUMO

UNLABELLED: The nicotine metabolite ratio (NMR), a stable measure of hepatic nicotine metabolism via the CYP2A6 pathway and total nicotine clearance, is a predictive biomarker of response to nicotine replacement therapy, with increased quit rates in slower metabolizers. Nicotine binds directly to nicotinic acetylcholine receptors (nAChRs) to exert its psychoactive effects. This study examined the relationship between NMR and nAChR (α4ß2* subtype) availability using PET imaging of the radiotracer 2-(18)F-fluoro-3-(2(S)-azetidinylmethoxy)pyridine (2-(18)F-FA-85380, or 2-(18)F-FA). METHODS: Twenty-four smokers-12 slow metabolizers (NMR < 0.26) and 12 normal metabolizers (NMR ≥ 0.26)-underwent 2-(18)F-FA-PET brain imaging after overnight nicotine abstinence (18 h before scanning), using a validated bolus-plus-infusion protocol. Availability of nAChRs was compared between NMR groups in a priori volumes of interest, with total distribution volume (VT/fP) being the measure of nAChR availability. Cravings to smoke were assessed before and after the scans. RESULTS: Thalamic nAChR α4ß2* availability was significantly reduced in slow nicotine metabolizers (P = 0.04). Slow metabolizers exhibited greater reductions in cravings after scanning than normal metabolizers; however, craving was unrelated to nAChR availability. CONCLUSION: The rate of nicotine metabolism is associated with thalamic nAChR availability. Additional studies could examine whether altered nAChR availability underlies the differences in treatment response between slow and normal metabolizers of nicotine.


Assuntos
Nicotina/metabolismo , Receptores Nicotínicos/metabolismo , Fumar/metabolismo , Adulto , Azetidinas , Disponibilidade Biológica , Encéfalo/metabolismo , Fissura , Feminino , Humanos , Infusões Intravenosas , Cinética , Espectroscopia de Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Nicotina/farmacocinética , Tomografia por Emissão de Pósitrons , Piridinas , Compostos Radiofarmacêuticos , Reprodutibilidade dos Testes , Fumar/psicologia , Tálamo/diagnóstico por imagem , Tabagismo/metabolismo , Tabagismo/psicologia , Adulto Jovem
16.
J Nucl Med ; 55(3): 500-507, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24525208

RESUMO

UNLABELLED: Tumor endothelial marker 1 (TEM1/endosialin) is a tumor vascular marker highly overexpressed in multiple human cancers with minimal expression in normal adult tissue. In this study, we report the preparation and evaluation of (124)I-MORAb-004, a humanized monoclonal antibody targeting an extracellular epitope of human TEM1 (hTEM1), for its ability to specifically and sensitively detect vascular cells expressing hTEM1 in vivo. METHODS: MORAb-004 was directly iodinated with (125)I and (124)I, and in vitro binding and internalization parameters were characterized. The in vivo behavior of radioiodinated MORAb-004 was characterized in mice bearing subcutaneous ID8 tumors enriched with mouse endothelial cells expressing hTEM1 and by biodistribution and small-animal immuno-PET studies. RESULTS: MORAb-004 was radiolabeled with high efficiency and isolated in high purity. In vitro studies demonstrated specific and sensitive binding of MORAb-004 to MS1 mouse endothelial cells expressing hTEM1, with no binding to control MS1 cells. (125)I-MORAb-004 and (124)I-MORAb-004 both had an immunoreactivity of approximately 90%. In vivo biodistribution experiments revealed rapid, highly specific and sensitive uptake of MORAb-004 in MS1-TEM1 tumors at 4 h (153.2 ± 22.2 percentage injected dose per gram [%ID/g]), 24 h (127.1 ± 42.9 %ID/g), 48 h (130.3 ± 32.4 %ID/g), 72 h (160.9 ± 32.1 %ID/g), and 6 d (10.7 ± 1.8 %ID/g). Excellent image contrast was observed with (124)I-immuno-PET. MORAb-004 uptake was statistically higher in TEM1-positive tumors than in control tumors. Binding specificity was confirmed by blocking studies using excess nonlabeled MORAb-004. CONCLUSION: In our preclinical model, with hTEM1 exclusively expressed on engineered murine endothelial cells that integrate into the tumor vasculature, (124)I-MORAb-004 displays high tumor-to-background tissue contrast for detection of hTEM1 in easily accessible tumor vascular compartments. These studies strongly suggest the clinical utility of (124)I-MORAb-004 immuno-PET in assessing TEM1 tumor-status.


Assuntos
Anticorpos Monoclonais , Antígenos CD/imunologia , Vasos Sanguíneos/metabolismo , Proteínas de Neoplasias/imunologia , Neoplasias/irrigação sanguínea , Tomografia por Emissão de Pósitrons/métodos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Linhagem Celular , Feminino , Humanos , Radioisótopos do Iodo , Marcação por Isótopo , Camundongos
17.
Nucl Med Biol ; 33(2): 203-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16546674

RESUMO

Overexpression of sigma-2 receptors in human tumors, such as melanoma, breast cancer, small cell lung carcinoma and prostate cancer, has been reported. Furthermore, the expression of sigma-2 receptors parallels the proliferative status of breast tumors implanted in nude mice. Thus, radiolabeled probes with a high affinity and high selectivity targeting sigma-2 receptors may be useful as tumor imaging agents. A conformationally flexible benzamide derivative, 5-bromo-2,3-dimethoxy-N-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-butyl]-benzamide, displayed greater than 1,000-fold selectivity for sigma-2 receptors (K(i)=8.2 and 12,900 nM for sigma-2 and sigma-1, respectively). The corresponding radioiodinated ligand, 5-iodo-2,3-dimethoxy-N-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-butyl]-benzamide ([(125)I]/[(123)I]I), was successfully prepared via an iododestannylation reaction. Binding studies carried out in membrane homogenates prepared from the mouse mammary tumor cell line (EMT6) with [(125)I]I showed the desired high binding affinity and high capacity (K(d)=0.68+/-0.06 nM, B(max)=1,005+/-46 fmol/mg protein). The sigma-2-like pharmacological profile of [(125)I]I binding sites was confirmed by competition studies. Similar binding parameters were also found in EMT6 xenografts (Day 10 and Day 19 implants) from BALB/c mice (K(d)=0.43-1.1 nM, B(max)=2,025-4,528 fmol/mg protein). It was determined by dissection and microSPECT imaging that [(125)I]/[(123)]I accumulated in EMT6 tumors established in BALB/c mice (Day 10 implants). Two hours after the tracer injection, the dissection ratio of EMT6 tumor to background (muscle) reached 6-7. However, microSPECT imaging could not clearly delineate the tumors, while the specific localization could be confirmed by ex vivo autoradiography. In summary, the novel iodinated ligand with high affinity and considerable selectivity for sigma-2 receptors may provide a useful tool to characterize sigma-2 receptors in vitro. Further modification of the ligand and the imaging parameters will be needed to improve the signal for in vivo detection.


Assuntos
Benzamidas/farmacocinética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/metabolismo , Receptores sigma/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Radioisótopos do Iodo/farmacocinética , Taxa de Depuração Metabólica , Camundongos , Especificidade de Órgãos , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
18.
Nucl Med Biol ; 31(5): 533-41, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15219270

RESUMO

A radioiodinated ligand, (R)-N-methyl-(2-[(125)I]iodo-phenoxy)-3-phenylpropylamine, [(125)I]2-INXT, targeting norepinephrine transporters (NET), was successfully prepared. A no-carrier-added product, [(125)I]2-INXT, displayed a saturable binding with a high affinity (K(d)=0.06 nM) in the homogenates prepared from rat cortical tissues as well as from LLC-PK(1) cells expressing NET. A relatively low number of binding sties (B(max)=55 fmol/mg protein) measured with [(125)I]2-INXT in rat cortical homogenates is consistent with the value reported for a known NET ligand, [(3)H]nisoxetine. Competition studies with various compounds on [(125)I]2-INXT binding clearly confirmed the pharmacological specificity and selectivity for NET binding sites. Following a tail-vein injection of [(125)I]2-INXT in rats, a good initial brain uptake was observed (0.56% dose at 2 min) followed by a slow washout from the brain (0.2% remained at 3 hours post-injection). The hypothalamus (a NET-rich region) to striatum (a region devoid of NET) ratio was 1.5 at 3 hours post-i.v. injection. Pretreatment of rats with nisoxetine significantly inhibited the uptake of [(125)I]2-INXT (70-100% inhibition) in locus coeruleus, hypothalamus and raphe nuclei, regions known to have a high density of NET; whereas escitalopram, a serotonin transporter ligand, did not show a similar effect. Ex vivo autoradiography of rat brain sections of [(125)I]2-INXT (at 3 hours after an i.v. injection) displayed an excellent regional brain localization pattern corroborated to the specific NET distribution in the brain. The specific brain localization was significantly reduced by a dose of nisoxetine pretreatment. Taken together, the data suggest that [(123)I]2-INXT may be useful for mapping NET binding sites in the brain.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Fluoxetina/análogos & derivados , Fluoxetina/farmacocinética , Simportadores/metabolismo , Animais , Células LLC-PK1 , Masculino , Taxa de Depuração Metabólica , Proteínas da Membrana Plasmática de Transporte de Norepinefrina , Especificidade de Órgãos , Ligação Proteica , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Suínos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA