Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Int J Hyperthermia ; 38(1): 650-662, 2021.
Article En | MEDLINE | ID: mdl-33882773

BACKGROUND: Metastatic prostate cancer in bone is difficult to treat as the tumor cells are relatively resistant to hormonal or chemotherapies when compared to primary prostate cancer. Irreversible electroporation (IRE) is a minimally invasive ablation procedure that has potential applications in the management of prostate cancer in bone. However, a common limitation of IRE is tumor recurrence, which arises from incomplete ablation that allows remaining cancer cells to proliferate. In this study, we combined IRE with radium-223 (Ra-223), a bone-seeking radionuclide that emits short track length alpha particles and thus is associated with reduced damage to the bone marrow and evaluated the impact of the combination treatment on bone-forming prostate cancer tumors. METHODS: The antitumor activity of IRE and Ra-223 as single agents and in combination was tested in vitro against three bone morphogenetic protein 4 (BMP4)-expressing prostate cancer cell lines (C4-2B-BMP4, Myc-CaP-BMP4, and TRAMP-C2-BMP4). Similar evaluation was performed in vivo using a bone-forming C4-2B-BMP4 tumor model in nude mice. RESULTS: IRE and Ra-223 as monotherapy inhibited prostate cancer cell proliferation in vitro, and their combination resulted in significant reduction in cell viability compared to monotherapy. In vivo evaluation revealed that IRE with single-dose administration of Ra-233, compared to IRE alone, reduced the rate of tumor recurrence by 40% following initial apparent complete ablation and decreased the rate of proliferation of incompletely ablated tumor as quantified in Ki-67 staining (53.58 ± 16.0% for IRE vs. 20.12 ± 1.63%; for IRE plus Ra-223; p = 0.004). Histological analysis qualitatively showed the enhanced killing of tumor cells adjacent to bone by Ra-223 compared to those treated with IRE alone. CONCLUSION: IRE in combination with Ra-223, which enhanced the destruction of cancer cells that are adjacent to bone, resulted in reduction of tumor recurrence through improved clearance of proliferative cells in the tumor region.


Prostatic Neoplasms , Radium , Animals , Electroporation , Humans , Male , Mice , Mice, Nude , Neoplasm Recurrence, Local , Prostatic Neoplasms/radiotherapy , Radium/therapeutic use
2.
Clin Cancer Res ; 27(11): 3253-3264, 2021 06 01.
Article En | MEDLINE | ID: mdl-33753455

PURPOSE: Radium-223 prolongs survival in a fraction of men with bone metastatic prostate cancer (PCa). However, there are no markers for monitoring response and resistance to Radium-223 treatment. Exosomes are mediators of intercellular communication and may reflect response of the bone microenvironment to Radium-223 treatment. We performed molecular profiling of exosomes and compared the molecular profile in patients with favorable and unfavorable overall survival. EXPERIMENTAL DESIGN: We performed exosomal transcriptome analysis in plasma derived from our preclinical models (MDA-PCa 118b tumors, TRAMP-C2/BMP4 PCa) and from the plasma of 25 patients (paired baseline and end of treatment) treated with Radium-223. All samples were run in duplicate, and array data analyzed with fold changes +2 to -2 and P < 0.05. RESULTS: We utilized the preclinical models to establish that genes derived from the tumor and the tumor-associated bone microenvironment (bTME) are differentially enriched in plasma exosomes upon Radium-223 treatment. The mouse transcriptome analysis revealed changes in bone-related and DNA damage repair-related pathways. Similar findings were observed in plasma-derived exosomes from patients treated with Radium-223 detected changes. In addition, exosomal transcripts detected immune-suppressors (e.g., PD-L1) that were associated with shorter survival to Radium-223. Treatment of the Myc-CaP mouse model with a combination of Radium-223 and immune checkpoint therapy (ICT) resulted in greater efficacy than monotherapy. CONCLUSIONS: These clinical and coclinical analyses showed that RNA profiling of plasma exosomes may be used for monitoring the bTME in response to treatment and that ICT may be used to increase the efficacy of Radium-223.


Bone Neoplasms/secondary , Extracellular Vesicles/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/therapeutic use , Radium/pharmacology , Radium/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Animals , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Cell Line, Tumor , Exosomes/genetics , Gene Expression Profiling , Humans , Male , Mice , Prostatic Neoplasms/mortality , RNA/genetics , Survival Rate
3.
Clin Cancer Res ; 26(22): 5830-5842, 2020 11 15.
Article En | MEDLINE | ID: mdl-32816889

PURPOSE: 90Y-FF-21101 is an Yttrium-90-conjugated, chimeric mAb that is highly specific for binding to human placental (P)-cadherin, a cell-to-cell adhesion molecule overexpressed and associated with cancer invasion and metastatic dissemination in many cancer types. We report the clinical activity of 90Y-FF-21101 in a first-in-human phase I study in patients with advanced solid tumors. PATIENTS AND METHODS: The safety and efficacy of 90Y-FF-21101 were evaluated in a phase I 3+3 dose-escalation study in patients with advanced solid tumors (n = 15) over a dose range of 5-25 mCi/m2. Dosimetry using 111In-FF-21101 was performed 1 week prior to assess radiation doses to critical organs. Patients who demonstrated clinical benefit received repeated 90Y-FF-21101 administration every 4 months. RESULTS: 111In-FF-21101 uptake was observed primarily in the spleen, kidneys, testes, lungs, and liver, with tumor uptake observed in the majority of patients. Organ dose estimates for all patients were below applicable limits. P-cadherin expression H-scores ranged from 0 to 242 with 40% of samples exhibiting scores ≥100. FF-21101 protein pharmacokinetics were linear with increasing antibody dose, and the mean half-life was 69.7 (±12.1) hours. Radioactivity clearance paralleled antibody clearance. A complete clinical response was observed in a patient with clear cell ovarian carcinoma, correlating with a high tumor P-cadherin expression. Stable disease was observed in a variety of other tumor types, without dose-limiting toxicity. CONCLUSIONS: The favorable safety profile and initial antitumor activity observed for 90Y-FF-21101 warrant further evaluation of this radioimmunotherapeutic (RIT) approach and provide initial clinical data supporting P-cadherin as a potential target for cancer treatment.


Antibodies, Monoclonal/administration & dosage , Cadherins/antagonists & inhibitors , Neoplasms/radiotherapy , Radioimmunotherapy , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Cadherins/genetics , Cadherins/immunology , Carcinoembryonic Antigen/genetics , Cell Adhesion/drug effects , Dose Fractionation, Radiation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoglobulins/immunology , Indium Radioisotopes/administration & dosage , Kidney/drug effects , Liver/drug effects , Lung/drug effects , Male , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Spleen/drug effects , Testis/drug effects , Yttrium Radioisotopes/administration & dosage
4.
Diagnostics (Basel) ; 10(3)2020 Feb 25.
Article En | MEDLINE | ID: mdl-32106426

Although 3'-deoxy-3'[(18)F]-fluorothymidine (FLT)- positron emission tomography (PET) has been utilized for tumor response assessment to neoadjuvant chemotherapy in soft tissue sarcomas, it has not been exploited for the assessment of early response to systematically targeted therapies. Herein, we investigated the 18F-FLT PET/CT kinetics in patients with sarcoma who received targeted therapies. Among 15 patients with sarcoma who underwent 18F-FLT PET/CT, 5 patients (33%) patients were imaged at three time points: At baseline and at 1-15 weeks (MDM2-inhibitor treatment), and 10 patients (67%) were imaged twice: At baseline and at 1-4 weeks (MDM2 inhibitor, n = 5 ;c-met inhibitor n = 5). The patients with sarcoma had a total of 18 identifiable tumors. Twelve of 15 patients (80%) demonstrated 18F-FLT concentrations changes early, i.e., at 1-4 weeks. Eight patients responded (53.3%), four patients progressed (26.7%) based on FLT change of more than 10% increase, and three patients (20%) demonstrated no change. 18F-FLT PET/CT may be used for early response imaging to molecularly targeted therapies in patients with sarcoma. Further larger studies in specific sarcoma sub-types are warranted.

5.
Diagnostics (Basel) ; 10(1)2020 Jan 06.
Article En | MEDLINE | ID: mdl-31935818

Although 2-deoxy-2-[18F]-fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) is a sensitive nuclear medicine modality, specificity for characterizing lung cancer is limited. Tumor proliferation and early response to molecularly targeted therapy could be visualized using 3'-deoxy-3'[(18)F]-fluorothymidine (18F-FLT) PET/CT. The superiority of 18F-FLT PET/CT over 18F-FDG PET/CT in early therapeutic monitoring has been well described in patients after chemotherapy, radiotherapy, and/or chemo/radiotherapy. In thispilot study, we explorethe use of 18F-FLT PET/CT as an early response evaluation modality in patients with lung cancerand provide specific case studies of patients with small cell lung cancer and non-small cell lung cancer who received novel targeted therapies. Early response for c-MET inhibitor was observed in four weeks and for MDM2 inhibitor in nine days.

6.
Appl Radiat Isot ; 155: 108936, 2020 Jan.
Article En | MEDLINE | ID: mdl-31655351

68Ga-PSMA-11 is currently one of the most investigated PET agents for imaging both recurrent prostate cancer and relevant metastases; however, the production and distribution of 68Ga-PSMA-11 is limited to a supply of only a few daily doses when using a commercially available 68Ge/68Ga generator. 68Ge/68Ga generators deliver only a modest amount of activity, up to 1850 MBq (50 mCi), when new, but it decreases with time. Additionally, the production of 68Ga/68Ge generators has not been able to meet the increasing demand of 68Ga radiotracers. In response to the need for a more economically viable alternative, the focus of this study was to provide a simple and efficient method for producing 68Ga-PSMA-11, using cyclotron-produced 68Ga that is ready for routine clinical practice.


Cyclotrons , Membrane Glycoproteins/chemistry , Organometallic Compounds/chemistry , Automation , Cell Line, Tumor , Gallium Isotopes , Gallium Radioisotopes , Humans , Male
7.
J Clin Endocrinol Metab ; 103(10): 3698-3705, 2018 10 01.
Article En | MEDLINE | ID: mdl-30032208

Context: Many differentiated thyroid cancers (DTC) dedifferentiate and become radioactive iodine (RAI)-refractory (RAIR) with worse outcomes. Targeted therapy (TTx) may downregulate MAPK signaling and sensitize tumors to RAI. Objective: We describe patients with RAIR DTC receiving TTx with demonstrated RAI uptake allowing for iodine-131 (I131) administration. Design: Charts of patients with metastatic, progressive, RAIR DTC in whom TTx increased RAI uptake on a diagnostic whole-body scan (WBS), were reviewed. Results of WBS, I131 administration, thyroglobulin (TG) panels, and cross-sectional studies were recorded. Setting: Thirteen patients [median age (range), 56 (45 to 75) years; seven men] were included; 11 (85%) had DTC, two (15%) had poorly DTC. Nine (69%) had BRAF mutations, three (23%) had RAS mutations, and one (8%) was wild type. Selective BRAF or an MEK inhibitor TTx was continued for a median (range) of 14.3 (1 to 76.4) months before diagnostic WBS. Results: Nine (69%) patients were treated with I131 [median (range) activity, 204.4 (150 to 253) mCi], after which TTx was discontinued. Median (range) follow-up was 8.3 (0 to 17.4) months after I131 therapy. All nine patients had durable disease control (three had partial response, six had stable disease). TG and TG antibody levels increased in patients who demonstrated uptake before TTx, and declined in eight of the nine patients after I131 treatment. Adverse events included pneumonitis and sialadenitis. Conclusion: TTx in BRAF-/RAS-mutated RAIR DTC resensitizes tumors to iodine. Subsequent I131 administration results in meaningful responses. Patient selection, adverse events, response duration, and survival impact require additional study.


Iodine Radioisotopes/therapeutic use , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/radiotherapy , Adenocarcinoma, Follicular/drug therapy , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/radiotherapy , Aged , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/pathology , Carcinoma, Papillary/radiotherapy , Cross-Sectional Studies , Female , Follow-Up Studies , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , Male , Middle Aged , Mutation , Prognosis , Prospective Studies , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Thyroglobulin/blood , Thyroid Neoplasms/pathology
8.
Sci Transl Med ; 8(367): 367ra167, 2016 11 30.
Article En | MEDLINE | ID: mdl-27903863

Targeting the androgen receptor (AR) pathway prolongs survival in patients with prostate cancer, but resistance rapidly develops. Understanding this resistance is confounded by a lack of noninvasive means to assess AR activity in vivo. We report intracellular accumulation of a secreted antigen-targeted antibody (SATA) that can be used to characterize disease, guide therapy, and monitor response. AR-regulated human kallikrein-related peptidase 2 (free hK2) is a prostate tissue-specific antigen produced in prostate cancer and androgen-stimulated breast cancer cells. Fluorescent and radio conjugates of 11B6, an antibody targeting free hK2, are internalized and noninvasively report AR pathway activity in metastatic and genetically engineered models of cancer development and treatment. Uptake is mediated by a mechanism involving the neonatal Fc receptor. Humanized 11B6, which has undergone toxicological tests in nonhuman primates, has the potential to improve patient management in these cancers. Furthermore, cell-specific SATA uptake may have a broader use for molecularly guided diagnosis and therapy in other cancers.


Antibodies/chemistry , Bone Neoplasms/diagnostic imaging , Histocompatibility Antigens Class I/chemistry , Prostatic Neoplasms/diagnostic imaging , Receptors, Androgen/chemistry , Receptors, Fc/chemistry , Tissue Kallikreins/chemistry , Adenocarcinoma/diagnostic imaging , Animals , Bone Neoplasms/secondary , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Phenotype , Positron-Emission Tomography , Prostatic Neoplasms/pathology , Tomography, X-Ray Computed , Treatment Outcome
9.
Nucl Med Biol ; 42(4): 375-80, 2015 Apr.
Article En | MEDLINE | ID: mdl-25577038

OBJECTIVES: Human tumors xenografted in immunodeficient mice are crucial models in nuclear medicine to evaluate the effectiveness of candidate diagnostic and therapeutic compounds. However, little attention has been focused on the biological profile of the host model and its potential effects on the bio-distribution and tumor targeting of the tracer compound under study. We specifically investigated the dissimilarity in bio-distribution of (111)In-DTPA-5A10, which targets free prostate specific antigen (fPSA), in two animal models. METHODS: In vivo bio-distribution studies of (111)In-DTPA-5A10 were performed in immunodeficient BALB/c-nu or NMRI-nu mice with subcutaneous (s.c.) LNCaP tumors. Targeting-specificity of the tracer was assessed by quantifying the uptake in (a) mice with s.c. xenografts of PSA-negative DU145 cells as well as (b) BALB/c-nu or NMRI-nu mice co-injected with an excess of non-labeled 5A10. Finally, the effect of neonatal Fc-receptor (FcRn) inhibition on the bio-distribution of the conjugate was studied by saturating FcRn-binding capacity with nonspecific IgG1. RESULTS: The inherent biological attributes of the mouse model substantially influenced the bio-distribution and pharmacokinetics of (111)In-DTPA-5A10. With LNCaP xenografts in BALB/c-nu mice (with intact B and NK cells but with deficient T cells) versus NMRI-nu mice (with intact B cells, increased NK cells and absent T cells), we observed a significantly higher hepatic accumulation (26 ± 3.9 versus 3.5 ± 0.4%IA/g respectively), and concomitantly lower tumor uptake (25 ± 11 versus 52 ± 10%IA/g respectively) in BALB/c-nu mice. Inhibiting FcRn by administration of nonspecific IgG1 just prior to (111)In-DTPA-5A10 did not change tumor accumulation significantly. CONCLUSIONS: We demonstrated that the choice of immunodeficient mouse model importantly influence the bio-distribution of (111)In-DTPA-5A10. This study further highlighted important considerations in the evaluation of preclinical tracers, with respect to gaining information on their performance in the translational setting. Investigators utilizing xenograft models need to assess not only radiolabeling strategies, but also the host immunological status.


Immunoconjugates/pharmacokinetics , Immunoglobulin G/chemistry , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Immunocompetence , Indium Radioisotopes , Isotope Labeling , Male , Mice , Pentetic Acid/chemistry , Prostatic Neoplasms/pathology , Tissue Distribution
10.
J Clin Invest ; 121(12): 4700-11, 2011 Dec.
Article En | MEDLINE | ID: mdl-22105174

Advanced human thyroid cancers, particularly those that are refractory to treatment with radioiodine (RAI), have a high prevalence of BRAF (v-raf murine sarcoma viral oncogene homolog B1) mutations. However, the degree to which these cancers are dependent on BRAF expression is still unclear. To address this question, we generated mice expressing one of the most commonly detected BRAF mutations in human papillary thyroid carcinomas (BRAF(V600E)) in thyroid follicular cells in a doxycycline-inducible (dox-inducible) manner. Upon dox induction of BRAF(V600E), the mice developed highly penetrant and poorly differentiated thyroid tumors. Discontinuation of dox extinguished BRAF(V600E) expression and reestablished thyroid follicular architecture and normal thyroid histology. Switching on BRAF(V600E) rapidly induced hypothyroidism and virtually abolished thyroid-specific gene expression and RAI incorporation, all of which were restored to near basal levels upon discontinuation of dox. Treatment of mice with these cancers with small molecule inhibitors of either MEK or mutant BRAF reduced their proliferative index and partially restored thyroid-specific gene expression. Strikingly, treatment with the MAPK pathway inhibitors rendered the tumor cells susceptible to a therapeutic dose of RAI. Our data show that thyroid tumors carrying BRAF(V600E) mutations are exquisitely dependent on the oncoprotein for viability and that genetic or pharmacological inhibition of its expression or activity is associated with tumor regression and restoration of RAI uptake in vivo in mice. These findings have potentially significant clinical ramifications.


Antineoplastic Agents/therapeutic use , Carcinoma, Papillary/drug therapy , Indoles/therapeutic use , Iodine Radioisotopes/pharmacokinetics , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/physiology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/physiology , Sulfonamides/therapeutic use , Thyroid Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Apoptosis/drug effects , Benzamides/administration & dosage , Benzamides/pharmacology , Benzamides/therapeutic use , Benzamides/toxicity , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , DNA Damage , Diphenylamine/administration & dosage , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Diphenylamine/toxicity , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Genes, Synthetic/drug effects , Humans , Indoles/administration & dosage , Indoles/pharmacology , Indoles/toxicity , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Signaling System/physiology , Mice , Mice, Transgenic , Mutation, Missense , Neoplasm Proteins/genetics , Point Mutation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfonamides/toxicity , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Vemurafenib
11.
Blood ; 118(18): 4817-28, 2011 Nov 03.
Article En | MEDLINE | ID: mdl-21849486

We report the findings from the first 10 patients with chemotherapy-refractory chronic lymphocytic leukemia (CLL) or relapsed B-cell acute lymphoblastic leukemia (ALL) we have enrolled for treatment with autologous T cells modified to express 19-28z, a second-generation chimeric antigen (Ag) receptor specific to the B-cell lineage Ag CD19. Eight of the 9 treated patients tolerated 19-28z(+) T-cell infusions well. Three of 4 evaluable patients with bulky CLL who received prior conditioning with cyclophosphamide exhibited either a significant reduction or a mixed response in lymphadenopathy without concomitant development of B-cell aplasia. In contrast, one patient with relapsed ALL who was treated in remission with a similar T-cell dose developed a predicted B-cell aplasia. The short-term persistence of infused T cells was enhanced by prior cyclophosphamide administration and inversely proportional to the peripheral blood tumor burden. Further analyses showed rapid trafficking of modified T cells to tumor and retained ex vivo cytotoxic potential of CD19-targeted T cells retrieved 8 days after infusion. We conclude that this adoptive T-cell approach is promising and more likely to show clinical benefit in the setting of prior conditioning chemotherapy and low tumor burden or minimal residual disease. These studies are registered at www.clinicaltrials.org as #NCT00466531 (CLL protocol) and #NCT01044069 (B-ALL protocol).


Antigens, CD19/immunology , Graft Survival , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/therapy , T-Lymphocytes/transplantation , Adult , Aged , Antigens, CD19/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/physiology , Female , Graft Survival/physiology , Humans , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/immunology , Male , Middle Aged , Recurrence , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Transplantation, Autologous , Treatment Failure
12.
Cancer Res ; 71(8): 2871-81, 2011 Apr 15.
Article En | MEDLINE | ID: mdl-21487038

Human T cells genetically modified to express chimeric antigen receptors (CAR) specific to the B cell tumor antigen CD19 can successfully eradicate systemic human CD19(+) tumors in immunocompromised SCID (severe combined immunodeficient)-Beige mice. However, in the clinical setting, CD4(+) CD25(hi) T regulatory cells (Treg) present within the tumor microenvironment may be potent suppressors of tumor-targeted effector T cells. In order to assess the impact of Tregs on CAR-modified T cells in the SCID-Beige xenotransplant model, we isolated, genetically targeted and expanded natural T regulatory cells (nTreg). In vitro nTregs modified to express CD19-targeted CARs efficiently inhibited the proliferation of activated human T cells, as well as the capacity of CD19-targeted 19-28z(+) effector T cells to lyse CD19(+) Raji tumor cells. Intravenous infusion of CD19-targeted nTregs into SCID-Beige mice with systemic Raji tumors traffic to sites of tumor and recapitulate a clinically relevant hostile tumor microenvironment. Antitumor efficacy of subsequently infused 19-28z(+) effector T cells was fully abrogated as assessed by long-term survival of treated mice. Optimal suppression by genetically targeted nTregs was dependent on nTreg to effector T-cell ratios and in vivo nTreg activation. Prior infusion of cyclophosphamide in the setting of this nTreg-mediated hostile microenvironment was able to restore the antitumor activity of subsequently infused 19-28z(+) effector T cells through the eradication of tumor-targeted nTregs. These findings have significant implications for the design of future clinical trials utilizing CAR-based adoptive T-cell therapies of cancer.


Antigens, CD19/immunology , Burkitt Lymphoma/therapy , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Receptors, Antigen/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/immunology , Cell Line, Tumor , Combined Modality Therapy , Cyclophosphamide/pharmacology , Cytotoxicity, Immunologic , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, SCID , NIH 3T3 Cells , Receptors, Antigen/biosynthesis , Receptors, Antigen/genetics , Retroviridae/genetics , Transduction, Genetic , Xenograft Model Antitumor Assays
13.
Blood ; 116(11): e18-25, 2010 Sep 16.
Article En | MEDLINE | ID: mdl-20511541

We have developed a dual bioluminescent reporter system allowing noninvasive, concomitant imaging of T-cell trafficking, expansion, and activation of nuclear factor of activated T cells (NFAT) in vivo. NFAT activation plays an important role in T-cell activation and T-cell development. Therefore we used this system to determine spatial-temporal activation patterns of (1) proliferating T lymphocytes during graft-versus-host disease (GVHD) and (2) T-cell precursors during T-cell development after allogeneic hematopoietic stem cell transplantation (HSCT). In the first days after HSCT, donor T cells migrated to the peripheral lymph nodes and the intestines, whereas the NFAT activation was dominant in the intestines, suggesting an important role for the intestines in the early stages of alloactivation during development of GVHD. After adoptive transfer of in vitro-derived T-cell receptor (TCR) H-Y transgenic T-cell precursors into B6 (H-2(b)) hosts of both sexes, NFAT signaling and development into CD4(+) or CD8(+) single-positive cells could only be detected in the thymus of female recipients indicating either absence of positive selection or prompt depletion of double-positive thymocytes in the male recipients. Because NFAT plays an important role in a wide range of cell types, our system could provide new insights into a variety of biologic processes.


Cell Movement , Cell Proliferation , Precursor Cells, T-Lymphoid/cytology , T-Lymphocytes/cytology , 3T3 Cells , Adoptive Transfer , Animals , Cell Line , Cell Line, Tumor , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Jurkat Cells , Lentivirus/genetics , Luciferases/genetics , Luciferases/metabolism , Luminescence , Luminescent Measurements/methods , Lymphocyte Activation , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , Precursor Cells, T-Lymphoid/metabolism , Precursor Cells, T-Lymphoid/transplantation , Promoter Regions, Genetic/genetics , T-Lymphocytes/metabolism
14.
J Nucl Med ; 51(1): 121-9, 2010 Jan.
Article En | MEDLINE | ID: mdl-20048131

UNLABELLED: Because of the recent development of an iodopyridopyrimidinone Abl protein kinase inhibitor (PKI), (124)I-SKI-212230 ((124)I-SKI230), we investigated the feasibility of a PET-based molecular imaging method for the direct visualization of Abl kinase expression and PKI treatment. METHODS: In vitro pharmacokinetic properties, including specific and nonspecific binding of (124)I-SKI230 to its Abl kinase target and interaction with other PKIs, were assessed in cell-free medium and chronic myelogenous leukemia (CML) cells overexpressing BCR-Abl (K562), in comparison with BT-474 cells that are low in Abl expression. In a xenograft tumor model, we assessed the in vivo pharmacokinetics of (124)I-SKI230 using PET and postmortem tissue sampling. We also tested a paradigm of (124)I-SKI230 PET after treatment of the animal with a dose of Abl-specific PKI for the monitoring of the tumor response. RESULTS: In vitro studies confirmed that SKI230 binds to Abl kinase with nanomolar affinity, that selective uptake occurs in cell lines known to express Abl kinase, that RNAi knock-down supports specificity of cellular uptake due to Abl kinase, and that imatinib, an archetype Abl PKI, completely displaces SKI230. With SKI230, we obtained successful in vivo PET of Abl-expressing human tumors in a nude rat. We were also able to demonstrate evidence of substrate inhibition of in vivo radiotracer uptake in the xenograft tumor after treatment of the animal as a model of PKI treatment monitoring. CONCLUSION: These results support the hypothesis that molecular imaging using PET will be useful for the study of in vivo pharmacodynamics of Abl PKI molecular therapy in humans.


Enzyme Inhibitors , Neoplasms/diagnostic imaging , Neoplasms/enzymology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyridones , Pyrimidines , Radiopharmaceuticals , Binding, Competitive/drug effects , Blotting, Western , Cell Line, Tumor , Enzyme Inhibitors/pharmacokinetics , Humans , Isotope Labeling , K562 Cells , Positron-Emission Tomography , Protein Binding , Pyridones/pharmacokinetics , Pyrimidines/pharmacokinetics , RNA, Small Interfering , Radiopharmaceuticals/pharmacokinetics
15.
Nat Med ; 15(3): 338-44, 2009 Mar.
Article En | MEDLINE | ID: mdl-19219023

We developed a new approach to bioluminescent T cell imaging using a membrane-anchored form of the Gaussia luciferase (GLuc) enzyme, termed extGLuc, which we could stably express in both mouse and human primary T cells. In vitro, extGLuc+ cells emitted significantly higher bioluminescent signal when compared to cells expressing GLuc, Renilla luciferase (RLuc) or membrane-anchored RLuc (extRLuc). In vivo, mouse extGLuc+ T cells showed higher bioluminescent signal when compared to GLuc+ and RLuc+ T cells. Application of this imaging approach to human T cells genetically modified to express tumor-specific chimeric antigen receptors (CARs) enabled us to show in vivo CAR-mediated T cell accumulation in tumor, T cell persistence over time and concomitant imaging of T cells and tumor cells modified to express firefly luciferase. This sensitive imaging technology has application to many in vivo cell-based studies in a wide array of mouse models.


Luciferases/genetics , T-Lymphocytes/cytology , Animals , Cell Membrane/enzymology , Coleoptera/enzymology , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Sensitivity and Specificity , Signal Transduction
16.
Cancer Res ; 67(23): 11463-9, 2007 Dec 01.
Article En | MEDLINE | ID: mdl-18056475

Activating mutations of BRAF occur in approximately 7% of all human tumors and in the majority of melanomas. These tumors are very sensitive to pharmacologic inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK), which causes loss of D-cyclin expression, hypophosphorylation of Rb, and G(1) arrest. Growth arrest is followed by differentiation or senescence and, in a subset of BRAF mutant tumors, by apoptosis. The former effects result in so-called "stable disease" and, in patients with cancer, can be difficult to distinguish from indolent tumor growth. The profound G(1) arrest induced by MEK inhibition in BRAF mutant tumors is associated with a marked decline in thymidine uptake and is therefore potentially detectable in vivo by noninvasive 3'-deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT) positron emission tomography (PET) imaging. In SKMEL-28 tumor xenografts, MEK inhibition completely inhibited tumor growth and induced differentiation with only modest tumor regression. MEK inhibition also resulted in a rapid decline in the [(18)F]FLT signal in V600E BRAF mutant SKMEL-28 xenografts but not in BRAF wild-type BT-474 xenografts. The data suggest that [(18)F]FLT PET can effectively image induction of G(1) arrest by MEK inhibitors in mutant BRAF tumors and may be a useful noninvasive method for assessing the early biological response to this class of drugs.


Benzamides/pharmacology , Diphenylamine/analogs & derivatives , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/diagnostic imaging , Melanoma/metabolism , Positron-Emission Tomography , Proto-Oncogene Proteins B-raf/genetics , Radiopharmaceuticals , Animals , Blotting, Western , Dideoxynucleosides/metabolism , Diphenylamine/pharmacology , Drug Resistance, Neoplasm , Female , G1 Phase/drug effects , G1 Phase/physiology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Melanoma/drug therapy , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/metabolism , Radiopharmaceuticals/pharmacokinetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
J Med Chem ; 50(23): 5853-7, 2007 Nov 15.
Article En | MEDLINE | ID: mdl-17956080

Tyrosine kinases often play pivotal roles in the pathogenesis of cancer and are good candidates for therapeutic intervention and targeted molecular imaging. The precursor synthesis, radiosynthesis, and biological characterization of a fluorine-18 analog of dasatinib, a multitargeted kinase inhibitor, are reported. Compound 5 potently inhibits Abl, Src, and Kit kinases and inhibits K562 and M07e/p210bcr-abl human leukemic cell growth. Using positron emission tomography, we visualized K562 tumor xenografts in mice with [18F]-5.


Fluorine Radioisotopes , Pyrimidines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Thiazoles/chemical synthesis , Animals , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Fusion Proteins, bcr-abl , Humans , Mice , Mice, Nude , Positron-Emission Tomography , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Transplantation, Heterologous , src-Family Kinases/antagonists & inhibitors
18.
Clin Cancer Res ; 13(18 Pt 1): 5426-35, 2007 Sep 15.
Article En | MEDLINE | ID: mdl-17855649

PURPOSE: Human T cells targeted to the B cell-specific CD19 antigen through retroviral-mediated transfer of a chimeric antigen receptor (CAR), termed 19z1, have shown significant but partial in vivo antitumor efficacy in a severe combined immunodeficient (SCID)-Beige systemic human acute lymphoblastic leukemia (NALM-6) tumor model. Here, we investigate the etiologies of treatment failure in this model and design approaches to enhance the efficacy of this adoptive strategy. EXPERIMENTAL DESIGN: A panel of modified CD19-targeted CARs designed to deliver combined activating and costimulatory signals to the T cell was generated and tested in vitro to identify an optimal second-generation CAR. Antitumor efficacy of T cells expressing this optimal costimulatory CAR, 19-28z, was analyzed in mice bearing systemic costimulatory ligand-deficient NALM-6 tumors. RESULTS: Expression of the 19-28z CAR, containing the signaling domain of the CD28 receptor, enhanced systemic T-cell antitumor activity when compared with 19z1 in treated mice. A treatment schedule of 4 weekly T-cell injections, designed to prolong in vivo T-cell function, further improved long-term survival. Bioluminescent imaging of tumor in treated mice failed to identify a conserved site of tumor relapse, consistent with successful homing by tumor-specific T cells to systemic sites of tumor involvement. CONCLUSIONS: Both in vivo costimulation and repeated administration enhance eradication of systemic tumor by genetically targeted T cells. The finding that modifications in CAR design as well as T-cell dosing allowed for the complete eradication of systemic disease affects the design of clinical trials using this treatment strategy.


Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/transplantation , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred Strains , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Immunologic/immunology , Recombinant Fusion Proteins/immunology , Retroviridae/genetics , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
19.
J Immunol ; 176(6): 3306-10, 2006 Mar 15.
Article En | MEDLINE | ID: mdl-16517696

Ligation of CD28 during Ag recognition plays an important role in the generation of effective T cell responses. However, its peripheral control of regulatory T cell function remains obscure. In this study, we show that naive wild-type or CD28(-/-) CD4(+)CD25(-) T cells exposed to peptide in vivo develop regulatory activity that suppresses the response of adoptively transferred naive T cells to a subsequent immunogenic challenge. We find that although CD28 is engaged during the initial peptide-priming event and is essential to sustain T cell survival, it is not sufficient to prevent the dominance of regulatory T cell function. Immunization with adjuvant abrogates regulatory dominance, reducing overall Foxp3 expression in a CD28-dependent manner. We conclude that CD28 licenses active immunization by regulating Ag-induced immunoregulation.


CD28 Antigens/immunology , T-Lymphocytes, Regulatory/immunology , Vaccination , Animals , CD28 Antigens/metabolism , Forkhead Transcription Factors/metabolism , Mice , Mice, Inbred BALB C , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism
20.
J Nucl Med ; 47(1): 140-9, 2006 Jan.
Article En | MEDLINE | ID: mdl-16391198

UNLABELLED: Multistep immune targeting holds great promise for radioimmunodiagnosis and therapy of cancer. Pretargeting of the tetrameric single-chain, variable-fragment streptavidin construct of the tetrameric monoclonal antibody CC49 with subsequent administration of radiolabeled 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA)-biotin has yielded promising results in TAG-72-expressing tumor xenograft models. A potential limitation of this approach, however, has been high and prolonged renal uptake of radioactivity. The objective of the current study, therefore, was to evaluate the reduction of kidney uptake of radiolabeled DOTA-biotin achieved by each of 4 different methods. METHODS: A human pancreatic adenocarcinoma xenograft model (HPAC) in nude mice was used. The animals were intravenously injected with the antibody-streptavidin construct and a synthetic clearing agent (biotinylated N-acetyl-galactosamine) 24 and 4 h, respectively, before the administration of (67)Ga-DOTA-biotin. For reduction of the renal uptake, different groups of mice were treated with streptavidin saturated with biotin, with several administrations of lysine or colchicine or with a succinylated antibody-streptavidin construct (resulting in a decreased electrical charge). All animals were sacrificed 24 h after injection of the (67)Ga-DOTA-biotin for biodistribution and quantitative autoradiography (QAR) studies and selected animals underwent microSPECT/microCT studies. RESULTS: There was marked targeting of the radiolabeled DOTA-biotin to tumor in all groups except in negative-control animals. Only succinylation of the scFv-CC49-streptavidin fusion protein significantly reduced ( approximately 30%) kidney uptake without affecting tumor activity. QAR corroborated these results and demonstrated that radiolabeled DOTA-biotin localized selectively in the renal cortex. Among the other experimental groups, there was no change in kidney uptake of the radiolabeled biotin. CONCLUSION: In contrast to directly labeled antibodies and antibody fragments, administration of the negatively charged amino acid lysine was largely ineffective in pretargeting strategies with a single-chain-immuno-streptavidin fusion protein. Succinylation of the scFv-CC49-streptavidin construct, on the other hand, reduces kidney uptake of subsequently administered radiolabeled biotin, presumably by inhibiting reuptake of the fusion protein in the proximal renal tubules, and, therefore, could significantly reduce renal doses and improve therapeutic indices associated with multistep immune targeting approaches to radioimmunotherapy.


Adenocarcinoma/metabolism , Adenocarcinoma/radiotherapy , Biotin/analogs & derivatives , Drug Delivery Systems/methods , Gallium Radioisotopes/pharmacokinetics , Kidney/metabolism , Organometallic Compounds/pharmacokinetics , Radioimmunotherapy/methods , Animals , Biotin/pharmacokinetics , Biotin/therapeutic use , Female , Gallium Radioisotopes/therapeutic use , Immunoglobulin Fragments/therapeutic use , Kidney/diagnostic imaging , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Organometallic Compounds/therapeutic use , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Radiotherapy Dosage , Streptavidin/pharmacokinetics , Streptavidin/therapeutic use , Tissue Distribution , Whole-Body Counting
...