Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
FASEB J ; 37(6): e22995, 2023 06.
Article in English | MEDLINE | ID: mdl-37219526

ABSTRACT

Immuno-oncology (IO)-based therapies such as checkpoint inhibitors, bi-specific antibodies, and CAR-T-cell therapies have shown significant success in the treatment of several cancer indications. However, these therapies can result in the development of severe adverse events, including cytokine release syndrome (CRS). Currently, there is a paucity of in vivo models that can evaluate dose-response relationships for both tumor control and CRS-related safety issues. We tested an in vivo PBMC humanized mouse model to assess both treatment efficacy against specific tumors and the concurrent cytokine release profiles for individual human donors after treatment with a CD19xCD3 bispecific T-cell engager (BiTE). Using this model, we evaluated tumor burden, T-cell activation, and cytokine release in response to bispecific T-cell-engaging antibody in humanized mice generated with different PBMC donors. The results show that PBMC engrafted NOD-scid Il2rgnull mice lacking expression of mouse MHC class I and II (NSG-MHC-DKO mice) and implanted with a tumor xenograft predict both efficacy for tumor control by CD19xCD3 BiTE and stimulated cytokine release. Moreover, our findings indicate that this PBMC-engrafted model captures variability among donors for tumor control and cytokine release following treatment. Tumor control and cytokine release were reproducible for the same PBMC donor in separate experiments. The PBMC humanized mouse model described here is a sensitive and reproducible platform that identifies specific patient/cancer/therapy combinations for treatment efficacy and development of complications.


Subject(s)
Leukocytes, Mononuclear , T-Lymphocytes , Humans , Animals , Mice , Mice, Inbred NOD , Treatment Outcome , Cytokine Release Syndrome , Cytokines , Disease Models, Animal , Mice, Knockout , Mice, SCID
2.
FASEB J ; 36(9): e22476, 2022 09.
Article in English | MEDLINE | ID: mdl-35959876

ABSTRACT

Human innate immunity plays a critical role in tumor surveillance and in immunoregulation within the tumor microenvironment. Natural killer (NK) cells are innate lymphoid cells that have opposing roles in the tumor microenvironment, including NK cell subsets that mediate tumor cell cytotoxicity and subsets with regulatory function that contribute to the tumor immune suppressive environment. The balance between effector and regulatory NK cell subsets has been studied extensively in murine models of cancer, but there is a paucity of models to study human NK cell function in tumorigenesis. Humanized mice are a powerful alternative to syngeneic mouse tumor models for the study of human immuno-oncology and have proven effective tools to test immunotherapies targeting T cells. However, human NK cell development and survival in humanized NOD-scid-IL2rgnull (NSG) mice are severely limited. To enhance NK cell development, we have developed NSG mice that constitutively expresses human Interleukin 15 (IL15), NSG-Tg(Hu-IL15). Following hematopoietic stem cell engraftment of NSG-Tg(Hu-IL15) mice, significantly higher levels of functional human CD56+ NK cells are detectable in blood and spleen, as compared to NSG mice. Hematopoietic stem cell (HSC)-engrafted NSG-Tg(Hu-IL15) mice also supported the development of human CD3+ T cells, CD20+ B cells, and CD33+ myeloid cells. Moreover, the growth kinetics of a patient-derived xenograft (PDX) melanoma were significantly delayed in HSC-engrafted NSG-Tg(Hu-IL15) mice as compared to HSC-engrafted NSG mice demonstrating that human NK cells have a key role in limiting the tumor growth. Together, these data demonstrate that HSC-engrafted NSG-Tg(Hu-IL15) mice support enhanced development of functional human NK cells, which limit the growth of PDX tumors.


Subject(s)
Immunity, Innate , Interleukin-15 , Animals , Disease Models, Animal , Humans , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-15/genetics , Killer Cells, Natural , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
3.
FASEB J ; 34(9): 12963-12975, 2020 09.
Article in English | MEDLINE | ID: mdl-32772418

ABSTRACT

Immunotherapy is a powerful treatment strategy being applied to cancer, autoimmune diseases, allergies, and transplantation. Although therapeutic monoclonal antibodies (mAbs) have demonstrated significant clinical efficacy, there is also the potential for severe adverse events, including cytokine release syndrome (CRS). CRS is characterized by the rapid production of inflammatory cytokines following delivery of therapy, with symptoms ranging from mild fever to life-threating pathology and multi-organ failure. Overall there is a paucity of models to reliably and accurately predict the induction of CRS by immune therapeutics. Here, we describe the development of a humanized mouse model based on the NOD-scid IL2rgnull (NSG) mouse to study CRS in vivo. PBMC-engrafted NSG, NSG-MHC-DKO, and NSG-SGM3 mice were used to study cytokine release in response to treatment with mAb immunotherapies. Our data show that therapeutic-stimulated cytokine release in these PBMC-based NSG models captures the variation in cytokine release between individual donors, is drug dependent, occurs in the absence of acute xeno-GVHD, highlighting the specificity of the assay, and shows a robust response following treatment with a TGN1412 analog, a CD28 superagonist. Overall our results demonstrate that PBMC-engrafted NSG models are rapid, sensitive, and reproducible platforms to screen novel therapeutics for CRS.


Subject(s)
Antibodies, Monoclonal/adverse effects , Cytokine Release Syndrome/immunology , Cytokines/immunology , Disease Models, Animal , Leukocytes, Mononuclear/immunology , Animals , Antibodies, Monoclonal/immunology , Cytokine Release Syndrome/chemically induced , Female , Mice , Mice, Inbred NOD , Mice, SCID
4.
FASEB J ; 32(3): 1537-1549, 2018 03.
Article in English | MEDLINE | ID: mdl-29146734

ABSTRACT

Establishment of an in vivo small animal model of human tumor and human immune system interaction would enable preclinical investigations into the mechanisms underlying cancer immunotherapy. To this end, nonobese diabetic (NOD).Cg- PrkdcscidIL2rgtm1Wjl/Sz (null; NSG) mice were transplanted with human (h)CD34+ hematopoietic progenitor and stem cells, which leads to the development of human hematopoietic and immune systems [humanized NSG (HuNSG)]. HuNSG mice received human leukocyte antigen partially matched tumor implants from patient-derived xenografts [PDX; non-small cell lung cancer (NSCLC), sarcoma, bladder cancer, and triple-negative breast cancer (TNBC)] or from a TNBC cell line-derived xenograft (CDX). Tumor growth curves were similar in HuNSG compared with nonhuman immune-engrafted NSG mice. Treatment with pembrolizumab, which targets programmed cell death protein 1, produced significant growth inhibition in both CDX and PDX tumors in HuNSG but not in NSG mice. Finally, inhibition of tumor growth was dependent on hCD8+ T cells, as demonstrated by antibody-mediated depletion. Thus, tumor-bearing HuNSG mice may represent an important, new model for preclinical immunotherapy research.-Wang, M., Yao, L.-C., Cheng, M., Cai, D., Martinek, J., Pan, C.-X., Shi, W., Ma, A.-H., De Vere White, R. W., Airhart, S., Liu, E. T., Banchereau, J., Brehm, M. A., Greiner, D. L., Shultz, L. D., Palucka, K., Keck, J. G. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/drug effects , Immunotherapy , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Neoplasms/immunology , Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
J Infect Dis ; 217(1): 58-63, 2017 12 27.
Article in English | MEDLINE | ID: mdl-29087482

ABSTRACT

Both Ebola virus (EBOV) and Reston virus (RESTV) cause disease in nonhuman primates, yet only EBOV causes disease in humans. To investigate differences in viral pathogenicity, humanized mice (hu-NSG-SGM3) were inoculated with EBOV or RESTV. Consistent with differences in disease in human infection, pronounced weight loss and markers of hepatic damage and disease were observed exclusively in EBOV-infected mice. These abnormalities were associated with significantly higher EBOV replication in the liver but not in the spleen, suggesting that in this model, efficiency of viral replication in select tissues early in infection may contribute to differences in viral pathogenicity.


Subject(s)
Ebolavirus/growth & development , Hemorrhagic Fever, Ebola/virology , Liver/virology , Virus Replication , Animals , Body Weight , Disease Models, Animal , Hemorrhagic Fever, Ebola/pathology , Humans , Liver Function Tests , Mice , Mice, SCID
6.
J Infect Dis ; 216(11): 1386-1397, 2017 12 12.
Article in English | MEDLINE | ID: mdl-28482001

ABSTRACT

Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne viral hemorrhagic disease seen exclusively in humans. Central nervous system (CNS) infection and neurological involvement have also been reported in CCHF. In the current study, we inoculated NSG-SGM3 mice engrafted with human hematopoietic CD34+ stem cells with low-passage CCHF virus strains isolated from human patients. In humanized mice, lethal disease develops, characterized by histopathological change in the liver and brain. To date, targets of neurological infection and disease have not been investigated in CCHF. CNS disease in humanized mice was characterized by gliosis, meningitis, and meningoencephalitis, and glial cells were identified as principal targets of infection. Humanized mice represent a novel lethal model for studies of CCHF countermeasures, and CCHF-associated CNS disease. Our data suggest a role for astrocyte dysfunction in neurological disease and identify key regions of infection in the CNS for future investigations of CCHF.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/pathogenicity , Hemorrhagic Fever, Crimean/pathology , Neuroglia/pathology , Neuroglia/virology , Animals , Antibodies, Viral , Brain/pathology , Cell Line , Central Nervous System/pathology , Central Nervous System/virology , Chlorocebus aethiops , Female , Gliosis/pathology , Gliosis/virology , Hematopoietic Stem Cell Transplantation , Hemorrhagic Fever, Crimean/virology , Humans , Liver/pathology , Meningitis/pathology , Meningitis/virology , Meningoencephalitis/pathology , Meningoencephalitis/virology , Mice , Tick-Borne Diseases/pathology , Vero Cells
7.
J Infect Dis ; 214(suppl 3): S308-S318, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27601621

ABSTRACT

The study of Ebola virus (EBOV) pathogenesis in vivo has been limited to nonhuman primate models or use of an adapted virus to cause disease in rodent models. Herein we describe wild-type EBOV (Makona variant) infection of mice engrafted with human hematopoietic CD34+ stem cells (Hu-NSG™-SGM3 mice; hereafter referred to as SGM3 HuMice). SGM3 HuMice support increased development of myeloid immune cells, which are primary EBOV targets. In SGM3 HuMice, EBOV replicated to high levels, and disease was observed following either intraperitoneal or intramuscular inoculation. Despite the high levels of viral antigen and inflammatory cell infiltration in the liver, the characteristic histopathology of Ebola virus disease was not observed, and this absence of severe immunopathology may have contributed to the recovery and survival of some of the animals. Future investigations into the underlying mechanisms of the atypical disease presentation in SGM3 HuMice will provide additional insights into the immunopathogenesis of severe EBOV disease.


Subject(s)
Antigens, Viral/immunology , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Animals , Disease Models, Animal , Ebolavirus/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/virology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Humans , Liver/immunology , Liver/pathology , Liver/virology , Lymphocytes/pathology , Lymphocytes/virology , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/pathology , Myeloid Cells/virology , Spleen/immunology , Spleen/pathology , Spleen/virology , Transgenes , Virus Replication
8.
Nat Rev Clin Oncol ; 20(3): 192-206, 2023 03.
Article in English | MEDLINE | ID: mdl-36635480

ABSTRACT

Immunotherapy has emerged as a promising treatment paradigm for many malignancies and is transforming the drug development landscape. Although immunotherapeutic agents have demonstrated clinical efficacy, they are associated with variable clinical responses, and substantial gaps remain in our understanding of their mechanisms of action and specific biomarkers of response. Currently, the number of preclinical models that faithfully recapitulate interactions between the human immune system and tumours and enable evaluation of human-specific immunotherapies in vivo is limited. Humanized mice, a term that refers to immunodeficient mice co-engrafted with human tumours and immune components, provide several advantages for immuno-oncology research. In this Review, we discuss the benefits and challenges of the currently available humanized mice, including specific interactions between engrafted human tumours and immune components, the development and survival of human innate immune populations in these mice, and approaches to study mice engrafted with matched patient tumours and immune cells. We highlight the latest advances in the generation of humanized mouse models, with the aim of providing a guide for their application to immuno-oncology studies with potential for clinical translation.


Subject(s)
Neoplasms , Animals , Mice , Humans , Neoplasms/therapy , Disease Models, Animal , Immunotherapy , Biomarkers , Immune System
9.
Lab Anim (NY) ; 52(7): 149-168, 2023 07.
Article in English | MEDLINE | ID: mdl-37386161

ABSTRACT

Humanized mouse models, created via transplantation of human hematopoietic tissues into immune-deficient mice, support a number of research applications, including transplantation immunology, virology and oncology studies. As an alternative to the bone marrow, liver, thymus humanized mouse, which uses fetal tissues for generating a chimeric human immune system, the NeoThy humanized mouse uses nonfetal tissue sources. Specifically, the NeoThy model incorporates hematopoietic stem and progenitor cells from umbilical cord blood (UCB) as well as thymus tissue that is typically discarded as medical waste during neonatal cardiac surgeries. Compared with fetal thymus tissue, the abundant quantity of neonatal thymus tissue offers the opportunity to prepare over 1,000 NeoThy mice from an individual thymus donor. Here we describe a protocol for processing of the neonatal tissues (thymus and UCB) and hematopoietic stem and progenitor cell separation, human leukocyte antigen typing and matching of allogenic thymus and UCB tissues, creation of NeoThy mice, assessment of human immune cell reconstitution and all experimental steps from planning and design to data analysis. This entire protocol takes a total of ~19 h to complete, with steps broken up into multiple sessions of 4 h or less that can be paused and completed over multiple days. The protocol can be completed, after practice, by individuals with intermediate laboratory and animal handling skills, enabling researchers to make effective use of this promising in vivo model of human immune function.


Subject(s)
Immune System , Thymus Gland , Humans , Animals , Mice , Disease Models, Animal , Liver , Research Personnel
10.
Blood ; 113(26): 6533-40, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19398716

ABSTRACT

Phase 1 testing of ezatiostat, a glutathione S-transferase P1-1 inhibitor, for the treatment of myelodysplastic syndrome was conducted in a multidose-escalation study. Patients received 10 dose levels (200, 400, 1000, 1400, 2000, 2400, 3000, 4000, 5000, and 6000 mg) of ezatiostat tablets in divided doses on days 1 to 7 of a 21-day cycle for a maximum of 8 cycles. The safety and pharmacokinetics of ezatiostat were evaluated. Forty-five patients with low to intermediate-2 International Prognostic Scoring System risk myelodysplastic syndrome were enrolled. No dose-limiting toxicities were observed. The most common grade 1 or 2, respectively, treatment-related adverse events were nonhematologic: nausea (56%, 9%), diarrhea (36%, 7%), vomiting (24%, 7%), abdominal pain (9%, 0%), constipation (4%, 9%), anorexia (3%, 7%), and dyspepsia (3%, 7%). Concentration of the primary active metabolite, TLK236, increased proportionate to ezatiostat dosage. Seventeen hematologic improvement (HI) responses by International Working Group criteria were observed at dose levels of 200 to 6000 mg/day with 11 HI responses at doses of 4000 to 6000 mg/day. HI responses occurred in all lineages including 3 bilineage and 1 complete cytogenetic response. Decreased number of red blood cell and platelet transfusions and in some cases transfusion independence were attained. Extended dose schedules of ezatiostat tablets are under investigation.


Subject(s)
Glutathione/analogs & derivatives , Myelodysplastic Syndromes/drug therapy , Prodrugs/therapeutic use , Abdominal Pain/chemically induced , Aged , Aged, 80 and over , Biotransformation , Dose-Response Relationship, Drug , Female , Fever/chemically induced , Food-Drug Interactions , Gastrointestinal Diseases/chemically induced , Glutathione/administration & dosage , Glutathione/adverse effects , Glutathione/pharmacokinetics , Glutathione/therapeutic use , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neutropenia/chemically induced , Prodrugs/administration & dosage , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Tablets
11.
Int J Gynecol Cancer ; 20(5): 772-80, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20973267

ABSTRACT

OBJECTIVE: To evaluate the safety and efficacy of canfosfamide in combination with pegylated liposomal doxorubicin (PLD) in platinum-resistant ovarian cancer (OC). METHODS: Patients with platinum-refractory or -resistant (primary or secondary) OC were randomized to receive canfosfamide at 1000 mg/m² and PLD at 50 mg/m² intravenously or PLD alone at 50 mg/m2 intravenously on day 1 every 28 days until tumor progression or unacceptable toxicity. The primary end point was progression-free survival (PFS). Other end points were objective response rate and safety. The study was originally planned for 244 patients. The trial was temporarily placed on hold after 125 patients were randomized while the results of another trial were being reviewed and the sponsor decided not to resume enrollment. The interim analysis became the final analysis. RESULTS: The median PFS was 5.6 months for canfosfamide + PLD (n = 65) versus 3.7 months for PLD (n = 60) (hazards ratio, 0.92; P = 0.7243). A preplanned subgroup analysis showed that 75 patients with platinum-refractory or primary platinum-resistant OC had a median PFS of 5.6 months for canfosfamide + PLD versus 2.9 months for PLD (hazards ratio, 0.55; P = 0.0425). Hematologic adverse events were 66% on the canfosfamide + PLD arm versus 44% on the PLD arm, manageable with dose reductions. Nonhematologic adverse events were similar for both arms. The incidence of palmar-plantar erythrodysesthesia and stomatitiswas lower on canfosfamide + PLD(23%, 31%, respectively) versus (39%, 49%, respectively) on PLD. CONCLUSIONS: Overall median PFS showed a positive trend but was not statistically significant. The median PFS in the platinum-refractory and primary platinum-resistant OC patients was significantly longer for canfosfamide + PLD versus PLD. Canfosfamide may ameliorate the palmar-plantar erythrodysesthesia and stomatitis known to be associated with PLD. Further study of this active well-tolerated regimen in platinum-refractory and primary platinum-resistant OC is planned. This study was registered at www.clinicaltrials.gov: NCT00350948.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/analogs & derivatives , Glutathione/analogs & derivatives , Ovarian Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/administration & dosage , Female , Glutathione/administration & dosage , Humans , Middle Aged , Platinum Compounds/therapeutic use , Survival Analysis , Treatment Outcome , Young Adult
12.
Methods Mol Biol ; 1953: 241-252, 2019.
Article in English | MEDLINE | ID: mdl-30912026

ABSTRACT

A significant obstacle to the study of human cancer biology and the testing of human specific immunotherapeutics is the paucity of translational models that recapitulate both the growth of human tumors and the functionality of human immune systems. Humanized mice engrafted with human hematopoietic stem cells (HSC) and patient-derived xenografts (PDX) enable preclinical investigation of the interactions between the human immune system and human cancer. We use immunodeficient non-obese diabetic (NOD, scid, gamma) NSG™ or NSG™-SGM3 mice as hosts for establishment of human immunity following HSC injection and for engraftment of human tumors. Here we describe a refined protocol for the subcutaneous implant of solid PDX tumors into humanized mice. Protocols to recover infiltrating immune cells from growing tumors and to evaluate the immune cell subsets by flow cytometry are also described.


Subject(s)
Neoplasm Transplantation/methods , Neoplasms/immunology , Transplantation, Heterologous/methods , Animals , Flow Cytometry/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunity , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology
13.
PLoS One ; 13(7): e0201104, 2018.
Article in English | MEDLINE | ID: mdl-30028878

ABSTRACT

Rift Valley fever (RVF) in humans is usually mild, but, in a subset of cases, can progress to severe hepatic and neurological disease. Rodent models of RVF generally develop acute severe clinical disease. Here, we inoculated humanized NSG-SGM3 mice with Rift Valley fever virus (RVFV) to investigate whether the presence of human immune cells in mice would alter the progression of RVFV infection to more closely model human disease. Despite increased human cytokine expression, including responses mirroring those seen in human disease, and decreased hepatic viral RNA levels at terminal euthanasia, both high- and low-dose RVFV inoculation resulted in lethal disease in all mice with comparable time-to-death as unengrafted mice.


Subject(s)
Hematopoietic Stem Cell Transplantation , Rift Valley Fever/immunology , Rift Valley Fever/therapy , Acute Disease , Animals , Antigens, CD34/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Humans , Liver/immunology , Liver/virology , Mice , RNA, Viral/metabolism , Rift Valley fever virus/physiology , Severity of Illness Index
14.
Mol Cancer Ther ; 17(5): 885-896, 2018 05.
Article in English | MEDLINE | ID: mdl-29483211

ABSTRACT

EGFR exon 20 insertions (Ex20Ins) account for 4% to 10% of EGFR activating mutations in non-small cell lung cancer (NSCLC). EGFR Ex20Ins tumors are generally unresponsive to first- and second-generation EGFR inhibitors, and current standard of care for NSCLC patients with EGFR Ex20Ins is conventional cytotoxic chemotherapy. Therefore, the development of an EGFR TKI that can more effectively target NSCLC with EGFR Ex20Ins mutations represents a major advance for this patient subset. Osimertinib is a third-generation EGFR TKI approved for the treatment of advanced NSCLC harboring EGFR T790M; however, the activity of osimertinib in EGFR Ex20Ins NSCLC has yet to be fully assessed. Using CRISPR-Cas 9 engineered cell lines carrying the most prevalent Ex20Ins mutations, namely Ex20Ins D770_N771InsSVD (22%) or Ex20Ins V769_D770InsASV (17%), and a series of patient-derived xenografts, we have characterized osimertinib and AZ5104 (a circulating metabolite of osimertinib) activities against NSCLC harboring Ex20Ins. We report that osimertinib and AZ5104 inhibit signaling pathways and cellular growth in Ex20Ins mutant cell lines in vitro and demonstrate sustained tumor growth inhibition of EGFR-mutant tumor xenograft harboring the most prevalent Ex20Ins in vivo The antitumor activity of osimertinib and AZ5104 in NSCLC harboring EGFR Ex20Ins is further described herein using a series of patient-derived xenograft models. Together these data support clinical testing of osimertinib in patients with EGFR Ex20Ins NSCLC. Mol Cancer Ther; 17(5); 885-96. ©2018 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Piperazines/pharmacology , Acrylamides , Aniline Compounds , Animals , COS Cells , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Chlorocebus aethiops , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Exons/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, SCID , Mutation , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays/methods
15.
Clin Cancer Res ; 10(11): 3689-98, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15173075

ABSTRACT

PURPOSE: To determine the dose-limiting toxicities, maximum tolerated dose, and pharmacokinetics of TLK286, a novel cancer prodrug, administered weekly. PATIENTS AND METHODS: Patients with advanced malignancies were treated with TLK286 administered weekly by i.v. infusion over 30 min in escalating doses 60-960 mg/m(2). A treatment cycle was defined as 3 weekly treatments. Patients underwent tumor assessments on day 43, and those patients receiving clinical benefit continued on treatment until disease progression or unacceptable toxicity. Safety was assessed by the WHO criteria. RESULTS: Thirty-seven patients received 111 cycles of TLK286 at eight dose levels (median, 3 cycles; range, 1-16 cycles). In this study, TLK286 given weekly at 960 mg/m(2) was well tolerated without dose-limiting toxicities. TLK286-related toxicities included grade 1-2 nausea and vomiting, fatigue and anemia. Nine of 31 evaluable patients continued therapy beyond day 43 and received a median of 5 cycles (range of 3-16 cycles) and experienced durable stable disease or minor tumor regression. Pharmacokinetic characteristics of TLK286 are described by an optimized two-compartment model. Mild to moderate renal or hepatic organ dysfunction did not impact the elimination of TLK286. CONCLUSIONS: TLK286 administered weekly at doses up to 960 mg/m(2) were well tolerated. The safety and antitumor activity observed in a broad range of cancer types supports Phase 2 disease-specific investigations of TLK286 given weekly at 960 mg/m(2).


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cytotoxins/therapeutic use , Glutathione/analogs & derivatives , Glutathione/administration & dosage , Glutathione/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Area Under Curve , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Models, Chemical , Neoplasm Metastasis , Time Factors
16.
Clin Cancer Res ; 9(5): 1628-38, 2003 May.
Article in English | MEDLINE | ID: mdl-12738715

ABSTRACT

PURPOSE: The purpose of this study was to determine the dose-limiting toxicities (DLTs), the maximum tolerated dose, and the pharmacokinetics of the novel glutathione analog TLK286 administered by i.v. infusion. EXPERIMENTAL DESIGN: Patients with advanced malignancies received i.v. TLK286 administered as a 30-min constant rate infusion once every 3 weeks in escalating doses from 60 to 1280 mg/m(2). Patients underwent tumor assessment on day 43 and continued on treatment until disease progression or unacceptable toxicity. RESULTS: A total of 35 patients were treated with 109 cycles of TLK286. At 1280 mg/m(2), 3 of 5 patients developed one of two observed dose limiting toxicities (DLTs). The DLTs were: mild pancreatitis (1 of 5) and bladder symptoms (2 of 5) consisting of hematuria, dysuria, and urinary frequency. All of the patients with DLTs continued on TLK286 treatment at 960 mg/m(2) (one dose below maximum tolerated dose) without recurrence of DLTs. DLTs were transient, resolved without sequelae, and noncumulative. TLK286-related toxicities included grade 1-2 nausea, vomiting, fatigue, transient microscopic hematuria, and anemia. Of 31 evaluable patients, 10 patients continued therapy (median six cycles; range, four to nine cycles). Pharmacokinetic studies of TLK286 on cycle 1 revealed a mean elimination half-life of 18 min (95% confidence interval, 16.1-19.9). Dose-proportional increases in both maximum blood concentrations and area under the blood-concentration-time curve were observed over the dose range of 60-960 mg/m(2). CONCLUSION: TLK286 was well tolerated in this study. TLK286 safety and pharmacokinetics support disease-specific evaluations of TLK286 at doses <1280 mg/m(2) administered once every three weeks in the treatment of patients with advanced malignancies.


Subject(s)
Cytotoxins/administration & dosage , Cytotoxins/pharmacokinetics , Glutathione/analogs & derivatives , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Adult , Aged , Cytotoxins/adverse effects , Dose-Response Relationship, Drug , Female , Glutathione/adverse effects , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasms/blood
17.
Clin Lung Cancer ; 16(3): 165-72, 2015 May.
Article in English | MEDLINE | ID: mdl-25838158

ABSTRACT

New approaches to optimization of cancer drug development in the laboratory and the clinic will be required to fully achieve the goal of individualized, precision cancer therapy. Improved preclinical models that more closely reflect the now recognized genomic complexity of human cancers are needed. Here we describe a collaborative research project that integrates core resources of The Jackson Laboratory Basic Science Cancer Center with genomics and clinical research facilities at the UC Davis Comprehensive Cancer Center to establish a clinically and genomically annotated patient-derived xenograft (PDX) platform designed to enhance new drug development and strategies for targeted therapies. Advanced stage non-small-cell lung cancer (NSCLC) was selected for initial studies because of emergence of a number of "druggable" molecular targets, and recent recognition of substantial inter- and intrapatient tumor heterogeneity. Additionally, clonal evolution after targeted therapy interventions make this tumor type ideal for investigation of this platform. Using the immunodeficient NOD scid gamma mouse, > 200 NSCLC tumor biopsies have been xenotransplanted. During the annotation process, patient tumors and subsequent PDXs are compared at multiple levels, including histomorphology, clinically applicable molecular biomarkers, global gene expression patterns, gene copy number variations, and DNA/chromosomal alterations. NSCLC PDXs are grouped into panels of interest according to oncogene subtype and/or histologic subtype. Multiregimen drug testing, paired with next-generation sequencing before and after therapy and timed tumor pharmacodynamics enables determination of efficacy, signaling pathway alterations, and mechanisms of sensitivity-resistance in individual models. This approach should facilitate derivation of new therapeutic strategies and the transition to individualized therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genomics , Lung Neoplasms/genetics , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID
18.
J Med Chem ; 47(20): 4875-80, 2004 Sep 23.
Article in English | MEDLINE | ID: mdl-15369391

ABSTRACT

We used protein affinity fingerprints to discover structurally novel inhibitors of cyclooxygenase-1 (COX-1) by screening a selected number of compounds, thus providing an alternative to extensive screening. From the affinity fingerprints of 19 known COX-1 inhibitors, a computational model for COX-1 inhibition was constructed and used to select candidate inhibitors from our compound library to be tested in the COX-1 assay. Subsequent refinement of the model by including affinity fingerprints of inactive compounds identified three molecules that were more potent than ibuprofen, a commonly used COX-1 inhibitor. These compounds are structurally distinct from those used to build the model and were discovered by testing only 62 library compounds. The discovery of these leads demonstrates the efficiency with which affinity fingerprints can identify novel bioactive chemotypes from known drugs.


Subject(s)
Combinatorial Chemistry Techniques/methods , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Models, Theoretical , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 1 , Drug Design , Drug Evaluation, Preclinical/methods , Ibuprofen/chemistry , Ibuprofen/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Prospective Studies , Prostaglandin-Endoperoxide Synthases , Quantitative Structure-Activity Relationship
19.
Anticancer Res ; 29(10): 3845-55, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19846918

ABSTRACT

BACKGROUND: DNA-damaging agents are widely used for the treatment of human malignancies. Agents containing the multifunctional alkylating moiety tetrakis(2-chloroethyl)phosphorodiamidic acid are currently under development as cancer therapeutics. MATERIALS AND METHODS: TLK58747, a phophorodiamidate-based prodrug, was tested in vivo for antitumor efficacy and safety. The in vitro responses of tumor cells to TLK58747 were examined by cytotoxicity assays, cell cycle analysis, immunoblots and microscopy. RESULTS: TLK58747 was efficacious in xenograft models of human breast, pancreas, and prostate cancer, as well as in leukemia and glioma. It caused less bone marrow suppression in rats than did cyclophosphamide. In vitro, TLK58747 inhibited the growth of a wide variety of cancer cells and activated the DNA damage-response pathway, leading to G(2)/M cell cycle arrest and subsequent premature senescence or apoptosis. CONCLUSION: TLK58747 is a promising new alkylating agent with broad antitumor activity and superior safety that warrants further development.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Damage , DNA, Neoplasm/drug effects , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents, Alkylating/toxicity , Cell Division/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , DNA, Neoplasm/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , G2 Phase/drug effects , HL-60 Cells , Humans , Male , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Organophosphorus Compounds/toxicity , Prodrugs/toxicity , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
20.
J Hematol Oncol ; 2: 20, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19439093

ABSTRACT

BACKGROUND: Ezatiostat hydrochloride liposomes for injection, a glutathione S-transferase P1-1 inhibitor, was evaluated in myelodysplastic syndrome (MDS). The objectives were to determine the safety, pharmacokinetics, and hematologic improvement (HI) rate. Phase 1-2a testing of ezatiostat for the treatment of MDS was conducted in a multidose-escalation, multicenter study. Phase 1 patients received ezatiostat at 5 dose levels (50, 100, 200, 400 and 600 mg/m2) intravenously (IV) on days 1 to 5 of a 14-day cycle until MDS progression or unacceptable toxicity. In phase 2, ezatiostat was administered on 2 dose schedules: 600 mg/m2 IV on days 1 to 5 or days 1 to 3 of a 21-day treatment cycle. RESULTS: 54 patients with histologically confirmed MDS were enrolled. The most common adverse events were grade 1 or 2, respectively, chills (11%, 9%), back pain (15%, 2%), flushing (19%, 0%), nausea (15%, 0%), bone pain (6%, 6%), fatigue (0%, 13%), extremity pain (7%, 4%), dyspnea (9%, 4%), and diarrhea (7%, 4%) related to acute infusional hypersensitivity reactions. The concentration of the primary active metabolites increased proportionate to ezatiostat dosage. Trilineage responses were observed in 4 of 16 patients (25%) with trilineage cytopenia. Hematologic Improvement-Erythroid (HI-E) was observed in 9 of 38 patients (24%), HI-Neutrophil in 11 of 26 patients (42%) and HI-Platelet in 12 of 24 patients (50%). These responses were accompanied by improvement in clinical symptoms and reductions in transfusion requirements. Improvement in bone marrow maturation and cellularity was also observed. CONCLUSION: Phase 2 studies of ezatiostat hydrochloride liposomes for injection in MDS are supported by the tolerability and HI responses observed. An oral formulation of ezatiostat hydrochloride tablets is also in phase 2 clinical development. TRIAL REGISTRATION: Clinicaltrials.gov: NCT00035867.


Subject(s)
Glutathione/analogs & derivatives , Myelodysplastic Syndromes/drug therapy , Prodrugs/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Dose-Response Relationship, Drug , Female , Glutathione/administration & dosage , Glutathione/adverse effects , Glutathione/chemistry , Humans , Hydrochloric Acid/administration & dosage , Hydrochloric Acid/chemistry , Injections , Liposomes , Male , Middle Aged , Prodrugs/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL