Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Blood ; 143(21): 2201-2216, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38447038

ABSTRACT

ABSTRACT: Fanconi anemia (FA) is an inherited DNA repair disorder characterized by bone marrow (BM) failure, developmental abnormalities, myelodysplasia, leukemia, and solid tumor predisposition. Allogeneic hematopoietic stem cell transplantation (allo-HSCT), a mainstay treatment, is limited by conditioning regimen-related toxicity and graft-versus-host disease (GVHD). Antibody-drug conjugates (ADCs) targeting hematopoietic stem cells (HSCs) can open marrow niches permitting donor stem cell alloengraftment. Here, we report that single dose anti-mouse CD45-targeted ADC (CD45-ADC) facilitated stable, multilineage chimerism in 3 distinct FA mouse models representing 90% of FA complementation groups. CD45-ADC profoundly depleted host stem cell enriched Lineage-Sca1+cKit+ cells within 48 hours. Fanca-/- recipients of minor-mismatched BM and single dose CD45-ADC had peripheral blood (PB) mean donor chimerism >90%; donor HSCs alloengraftment was verified in secondary recipients. In Fancc-/- and Fancg-/- recipients of fully allogeneic grafts, PB mean donor chimerism was 60% to 80% and 70% to 80%, respectively. The mean percent donor chimerism in BM and spleen mirrored PB results. CD45-ADC-conditioned mice did not have clinical toxicity. A transient <2.5-fold increase in hepatocellular enzymes and mild-to-moderate histopathological changes were seen. Under GVHD allo-HSCT conditions, wild-type and Fanca-/- recipients of CD45-ADC had markedly reduced GVHD lethality compared with lethal irradiation. Moreover, single dose anti-human CD45-ADC given to rhesus macaque nonhuman primates on days -6 or -10 was at least as myeloablative as lethal irradiation. These data suggest that CD45-ADC can potently promote donor alloengraftment and hematopoiesis without significant toxicity or severe GVHD, as seen with lethal irradiation, providing strong support for clinical trial considerations in highly vulnerable patients with FA.


Subject(s)
Fanconi Anemia , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunoconjugates , Leukocyte Common Antigens , Animals , Fanconi Anemia/therapy , Mice , Graft vs Host Disease/pathology , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Transplantation Conditioning/methods , Transplantation, Homologous , Mice, Inbred C57BL , Mice, Knockout
2.
J Allergy Clin Immunol ; 153(1): 341-348.e3, 2024 01.
Article in English | MEDLINE | ID: mdl-37567393

ABSTRACT

BACKGROUND: Mutations in the recombinase-activating genes 1 and 2 (RAG1, RAG2) cause a spectrum of phenotypes, ranging from severe combined immune deficiency to combined immune deficiency with immune dysregulation (CID-ID). Hematopoietic cell transplantation is a curative option. Use of conditioning facilitates robust and durable stem cell engraftment and immune reconstitution but may cause toxicity. Transplantation from haploidentical donors is associated with poor outcome in patients with CID-ID. OBJECTIVES: We sought to evaluate multilineage engraftment and immune reconstitution after conditioning with CD45-antibody drug conjugate (CD45-ADC) as a single agent in hypomorphic mice with Rag1 mutation treated with congenic and haploidentical hematopoietic cell transplantation. METHODS: Rag1-F971L mice, a model of CID-ID, were conditioned with various doses of CD45-ADC, total body irradiation, or isotype-ADC, and then given transplants of total bone marrow cells from congenic or haploidentical donors. Flow cytometry was used to assess chimerism and immune reconstitution. Histology was used to document reconstitution of thymic architecture. RESULTS: Conditioning with CD45-ADC as a single agent allowed robust engraftment and immune reconstitution, with restoration of thymus, bone marrow, and peripheral compartments. The optimal doses of CD45-ADC were 1.5 mg/kg and 5 mg/kg for congenic and haploidentical transplantation, respectively. No graft-versus-host disease was observed. CONCLUSIONS: Conditioning with CD45-ADC alone allows full donor chimerism and immune reconstitution in Rag1 hypomorphic mice even following haploidentical transplantation, opening the way for the implementation of similar approaches in humans.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes , Humans , Mice , Animals , Transplantation Conditioning , Bone Marrow Transplantation , Immunologic Deficiency Syndromes/therapy , Homeodomain Proteins/genetics
3.
Blood ; 139(11): 1743-1759, 2022 03 17.
Article in English | MEDLINE | ID: mdl-34986233

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment of patients with nonmalignant or malignant blood disorders. Its success has been limited by graft-versus-host disease (GVHD). Current systemic nontargeted conditioning regimens mediate tissue injury and potentially incite and amplify GVHD, limiting the use of this potentially curative treatment beyond malignant disorders. Minimizing systemic nontargeted conditioning while achieving alloengraftment without global immune suppression is highly desirable. Antibody-drug-conjugates (ADCs) targeting hematopoietic cells can specifically deplete host stem and immune cells and enable alloengraftment. We report an anti-mouse CD45-targeted-ADC (CD45-ADC) that facilitates stable murine multilineage donor cell engraftment. Conditioning with CD45-ADC (3 mg/kg) was effective as a single agent in both congenic and minor-mismatch transplant models resulting in full donor chimerism comparable to lethal total body irradiation (TBI). In an MHC-disparate allo-HSCT model, pretransplant CD45-ADC (3 mg/kg) combined with low-dose TBI (150 cGy) and a short course of costimulatory blockade with anti-CD40 ligand antibody enabled 89% of recipients to achieve stable alloengraftment (mean value: 72%). When CD45-ADC was combined with pretransplant TBI (50 cGy) and posttransplant rapamycin, cyclophosphamide (Cytoxan), or a JAK inhibitor, 90% to 100% of recipients achieved stable chimerism (mean: 77%, 59%, 78%, respectively). At a higher dose (5 mg/kg), CD45-ADC as a single agent was sufficient for rapid, high-level multilineage chimerism sustained through the 22 weeks observation period. Therefore, CD45-ADC has the potential utility to confer the benefit of fully myeloablative conditioning but with substantially reduced toxicity when given as a single agent or at lower doses in conjunction with reduced-intensity conditioning.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunoconjugates , Animals , Chimerism , Hematopoietic Stem Cell Transplantation/adverse effects , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/toxicity , Mice , Transplantation Conditioning/methods
4.
J Allergy Clin Immunol ; 147(1): 309-320.e6, 2021 01.
Article in English | MEDLINE | ID: mdl-32387109

ABSTRACT

BACKGROUND: Mutations in the recombinase-activating genes cause severe immunodeficiency, with a spectrum of phenotypes ranging from severe combined immunodeficiency to immune dysregulation. Hematopoietic stem cell transplantation is the only curative option, but a high risk of graft failure and poor immune reconstitution have been observed in the absence of myeloablation. OBJECTIVES: Our aim was to improve multilineage engraftment; we tested nongenotoxic conditioning with anti-CD45 mAbs conjugated with saporin CD45 (CD45-SAP). METHODS: Rag1-KO and Rag1-F971L mice, which represent models of severe combined immune deficiency and combined immune deficiency with immune dysregulation, respectively, were conditioned with CD45-SAP, CD45-SAP plus 2 Gy of total body irradiation (TBI), 2 Gy of TBI, 8 Gy of TBI, or no conditioning and treated by using transplantation with lineage-negative bone marrow cells from wild-type mice. Flow cytometry and immunohistochemistry were used to assess engraftment and immune reconstitution. Antibody responses to 2,4,6-trinitrophenyl-conjugated keyhole limpet hemocyanin were measured by ELISA, and presence of autoantibody was detected by microarray. RESULTS: Conditioning with CD45-SAP enabled high levels of multilineage engraftment in both Rag1 mutant models, allowed overcoming of B- and T-cell differentiation blocks and thymic epithelial cell defects, and induced robust cellular and humoral immunity in the periphery. CONCLUSIONS: Conditioning with CD45-SAP allows multilineage engraftment and robust immune reconstitution in mice with either null or hypomorphic Rag mutations while preserving thymic epithelial cell homeostasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow Transplantation , Homeodomain Proteins/genetics , Immunoconjugates/pharmacology , Leukocyte Common Antigens/antagonists & inhibitors , Saporins/pharmacology , Severe Combined Immunodeficiency/therapy , Transplantation Conditioning , Allografts , Animals , Antibodies, Monoclonal/adverse effects , Homeodomain Proteins/immunology , Immunoconjugates/adverse effects , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Mice , Mice, Knockout , Saporins/adverse effects , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology
5.
Blood ; 124(19): 2937-47, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25202142

ABSTRACT

The glycosyltransferase gene, Ext1, is essential for heparan sulfate production. Induced deletion of Ext1 selectively in Mx1-expressing bone marrow (BM) stromal cells, a known population of skeletal stem/progenitor cells, in adult mice resulted in marked changes in hematopoietic stem and progenitor cell (HSPC) localization. HSPC egressed from BM to spleen after Ext1 deletion. This was associated with altered signaling in the stromal cells and with reduced vascular cell adhesion molecule 1 production by them. Further, pharmacologic inhibition of heparan sulfate mobilized qualitatively more potent and quantitatively more HSPC from the BM than granulocyte colony-stimulating factor alone, including in a setting of granulocyte colony-stimulating factor resistance. The reduced presence of endogenous HSPC after Ext1 deletion was associated with engraftment of transfused HSPC without any toxic conditioning of the host. Therefore, inhibiting heparan sulfate production may provide a means for avoiding the toxicities of radiation or chemotherapy in HSPC transplantation for nonmalignant conditions.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Heparitin Sulfate/biosynthesis , N-Acetylglucosaminyltransferases/metabolism , Stromal Cells/metabolism , Transplantation Conditioning , Animals , Anticoagulants/pharmacology , Binding, Competitive/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Green Fluorescent Proteins/genetics , Heparin/pharmacology , Heparitin Sulfate/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , N-Acetylglucosaminyltransferases/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Stromal Cells/immunology , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
7.
bioRxiv ; 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37732224

ABSTRACT

Tissue resident myeloid cells (TRM) in adults have highly variable lifespans and may be derived from early embryonic yolk sac, fetal liver or bone marrow. Some of these TRM are known pathogenic participants in congenital and acquired diseases. Myeloablative conditioning and hematopoietic stem cell transplant can replace long-lived brain TRM resulting in clinical improvements in metabolic storage diseases. With the advent of antibody-drug-conjugate (ADC) targeted cell killing as a cell selective means of transplant conditioning, we assessed the impact of anti-CD45-ADC on TRM in multiple tissues. Replacement of TRM ranged from 40 to 95 percent efficiencies in liver, lung, and skin tissues, after a single anti-CD45-ADC dose and bone marrow hematopoietic cell transfer. Of note, the population size of TRM in tissues returned to pre-treatment levels suggesting a regulated control of TRM abundance. As expected, brain, microglia were not affected, but brain monocytes and macrophages were 50% replaced. Anti-CD45-ADC and adoptive cell transfer were then tested in the chronic acquired condition, atherosclerosis exacerbated by Tet2 mutant clonal hematopoiesis. Plaque resident myeloid cells were efficiently replaced with anti-CD45-ADC and wild-type bone marrow cells. Notably, this reduced existent atherosclerotic plaque burden. Overall, these results indicate that anti-CD45-ADC clears both HSC and TRM niches enabling cell replacement to achieve disease modification in a resident myeloid cell driven disease.

8.
Blood Adv ; 7(22): 6964-6973, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37748049

ABSTRACT

Tissue-resident myeloid (TRM) cells in adults have highly variable lifespans, and may be derived from early embryonic yolk sac, fetal liver, or bone marrow. Some of these TRM cells are known pathogenic participants in congenital and acquired diseases. Myeloablative conditioning and hematopoietic stem cell transplantation can replace long-lived brain TRM cells, resulting in clinical improvements in metabolic storage diseases. With the advent of antibody-drug conjugate (ADC)-targeted cell killing as a cell-selective means of transplant conditioning, we assessed the impact of anti-CD45-ADC on TRM cells in multiple tissues. Replacement of TRM cells ranged from 40% to 95% efficiencies in liver, lung, and skin tissues, after a single anti-CD45-ADC dose and bone marrow hematopoietic cell transfer. Of note, the population size of TRM cells in tissues returned to pretreatment levels, suggesting a regulated control of TRM cell abundance. As expected, brain microglia were not affected, but brain monocytes and macrophages were 50% replaced. Anti-CD45-ADC and adoptive cell transfer were then tested in the chronic acquired condition, atherosclerosis exacerbated by Tet2 mutant clonal hematopoiesis. Plaque-resident myeloid cells were efficiently replaced with anti-CD45-ADC and wild-type bone marrow cells. Notably, this reduced existent atherosclerotic plaque burden. Overall, these results indicate that the anti-CD45-ADC clears both hematopoietic stem and TRM cells from their niches, enabling cell replacement to achieve disease modification in a resident myeloid cell-driven disease.


Subject(s)
Immunoconjugates , Adult , Humans , Immunoconjugates/pharmacology , Macrophages , Monocytes , Bone Marrow , Microglia
9.
Nat Commun ; 14(1): 6291, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828021

ABSTRACT

Hematopoietic stem cell (HSC) gene therapy has curative potential; however, its use is limited by the morbidity and mortality associated with current chemotherapy-based conditioning. Targeted conditioning using antibody-drug conjugates (ADC) holds promise for reduced toxicity in HSC gene therapy. Here we test the ability of an antibody-drug conjugate targeting CD117 (CD117-ADC) to enable engraftment in a non-human primate lentiviral gene therapy model of hemoglobinopathies. Following single-dose CD117-ADC, a >99% depletion of bone marrow CD34 + CD90 + CD45RA- cells without lymphocyte reduction is observed, which results are not inferior to multi-day myeloablative busulfan conditioning. CD117-ADC, similarly to busulfan, allows efficient engraftment, gene marking, and vector-derived fetal hemoglobin induction. Importantly, ADC treatment is associated with minimal toxicity, and CD117-ADC-conditioned animals maintain fertility. In contrast, busulfan treatment commonly causes severe toxicities and infertility in humans. Thus, the myeloablative capacity of single-dose CD117-ADC is sufficient for efficient engraftment of gene-modified HSCs while preserving fertility and reducing adverse effects related to toxicity in non-human primates. This targeted conditioning approach thus provides the proof-of-principle to improve risk-benefit ratio in a variety of HSC-based gene therapy products in humans.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunoconjugates , Animals , Busulfan/pharmacology , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells , Immunoconjugates/pharmacology , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/therapeutic use , Macaca mulatta/immunology
10.
Mol Pharm ; 9(5): 1425-34, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22486564

ABSTRACT

The evasion of apoptosis is a key characteristic of cancer, and thus strategies to selectively induce apoptosis in cancer cells hold considerable promise in personalized anticancer therapy. Structurally similar procaspase activating compounds PAC-1 and S-PAC-1 restore procaspase-3 activity through the chelation of inhibitory zinc ions in vitro, induce apoptotic death of cancer cells in culture, and reduce tumor burden in vivo. Ip or iv administrations of high doses of PAC-1 are transiently neurotoxic in vivo, while S-PAC-1 is safe even at very high doses and has been evaluated in a phase I clinical trial of pet dogs with spontaneously occurring lymphoma. Here we show that PAC-1 and S-PAC-1 have similar mechanisms of cell death induction at low concentrations (less than 50 µM), but at high concentrations PAC-1 displays unique cell death induction features. Cells treated with a high concentration of PAC-1 have a distinctive gene expression profile, unusual cellular and mitochondrial morphology, and an altered intracellular Ca(2+) concentration, indicative of endoplasmic reticulum (ER) stress-induced apoptosis. These studies suggest strategies for anticancer clinical development, specifically bolus dosing for PAC-1 and continuous rate infusion for S-PAC-1.


Subject(s)
Caspase 3/metabolism , Cell Death/drug effects , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Calcium/metabolism , Cell Line, Tumor , Dogs , Female , HL-60 Cells , HeLa Cells , Humans , Hydrazones/pharmacology , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Piperazines/pharmacology , Zinc/metabolism
11.
Invest New Drugs ; 29(4): 562-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20169400

ABSTRACT

Cribrostatin 6 is a quinone-containing natural product that induces the death of cancer cell lines in culture, and its mechanism of action and scope of activity are unknown. Here we show that cribrostatin 6 has broad anticancer activity, potently inducing apoptotic cell death that is not preceded by any defined cell cycle arrest. Consistent with this data, we find that cribrostatin 6 treated cells have large amounts of reactive oxygen species (ROS) and, based on transcript profiling experiments and other data, this ROS generation is likely the primary mechanism by which cribrostatin 6 induces apoptosis. Given the success of certain ROS producers as anticancer agents, cribrostatin 6 has potential as a novel chemotherapeutic agent.


Subject(s)
Isoquinolines/pharmacology , Reactive Oxygen Species/metabolism , 3T3 Cells , Acetylcysteine/pharmacology , Animals , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoprotection/drug effects , DNA/metabolism , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Ethidium/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Isoquinolines/chemical synthesis , Isoquinolines/chemistry , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects
12.
J Am Chem Soc ; 132(15): 5469-78, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20345134

ABSTRACT

Deoxynyboquinone (DNQ) is a potent antineoplastic agent with an unknown mechanism of action. Here we describe a facile synthetic route to this anthraquinone, and we use this material to determine the mechanism by which DNQ induces death in cancer cells. DNQ was synthesized in seven linear steps through a route employing three palladium-mediated coupling reactions. Experiments performed on cancer cells grown in hypoxia and normoxia strongly suggest that DNQ undergoes bioreduction to its semiquinone, which then is re-oxidized by molecular oxygen, forming superoxide that induces cell death. Furthermore, global transcript profiling of cells treated with DNQ shows elevation of transcripts related to oxidative stress, a result confirmed at the protein level by Western blotting. In contrast to most other antineoplastic agents that generate reactive oxygen species (ROS), DNQ potently induces death of cancer cells in culture, with IC(50) values between 16 and 210 nM. In addition, unlike the experimental therapeutic elesclomol, DNQ is still able to induce cancer cell death under hypoxic conditions. This mechanistic understanding of DNQ will allow for a more comprehensive evaluation of the potential of direct ROS generation as an anticancer strategy, and DNQ itself has potential as a novel anticancer agent.


Subject(s)
Cell Death/drug effects , Quinones/chemical synthesis , Quinones/toxicity , Aza Compounds/chemical synthesis , Aza Compounds/toxicity , Cell Cycle/drug effects , Cell Line, Tumor , HeLa Cells , Heme Oxygenase-1/metabolism , Humans , Reactive Oxygen Species/metabolism , Topoisomerase II Inhibitors
13.
Mol Ther Methods Clin Dev ; 17: 455-464, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32226796

ABSTRACT

Conditioning chemotherapy is used to deplete hematopoietic stem cells in the recipient's marrow, facilitating donor cell engraftment. Although effective, a major issue with chemotherapy is the systemic genotoxicity that increases the risk for secondary malignancies. Antibody conjugates targeting hematopoietic cells are an emerging non-genotoxic method of opening the marrow niche and promoting engraftment of transplanted cells while maintaining intact marrow cellularity. Specifically, this platform would be useful in diseases associated with DNA damage or cancer predisposition, such as dyskeratosis congenita, Schwachman-Diamond syndrome, and Fanconi anemia (FA). Our approach utilizes antibody-drug conjugates (ADC) as an alternative conditioning regimen in an FA mouse model of autologous transplantation. Antibodies targeting either CD45 or CD117 were conjugated to saporin (SAP), a ribosomal toxin. FANCA knockout mice were conditioned with either CD45-SAP or CD117-SAP prior to receiving whole marrow from a heterozygous healthy donor. Bone marrow and peripheral blood analysis revealed equivalent levels of donor engraftment, with minimal toxicity in ADC-treated groups as compared with cyclophosphamide-treated controls. Our findings suggest ADCs may be an effective conditioning strategy in stem cell transplantation not only for diseases where traditional chemotherapy is not tolerated, but also more broadly for the field of blood and marrow transplantation.

14.
Nat Commun ; 10(1): 617, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30728354

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is a curative therapy for blood and immune diseases with potential for many settings beyond current standard-of-care. Broad HSCT application is currently precluded largely due to morbidity and mortality associated with genotoxic irradiation or chemotherapy conditioning. Here we show that a single dose of a CD117-antibody-drug-conjugate (CD117-ADC) to saporin leads to > 99% depletion of host HSCs, enabling rapid and efficient donor hematopoietic cell engraftment. Importantly, CD117-ADC selectively targets hematopoietic stem cells yet does not cause clinically significant side-effects. Blood counts and immune cell function are preserved following CD117-ADC treatment, with effective responses by recipients to both viral and fungal challenges. These results suggest that CD117-ADC-mediated HSCT pre-treatment could serve as a non-myeloablative conditioning strategy for the treatment of a wide range of non-malignant and malignant diseases, and might be especially suited to gene therapy and gene editing settings in which preservation of immunity is desired.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Immunoconjugates/pharmacology , Proto-Oncogene Proteins c-kit/immunology , Animals , Bone Marrow/drug effects , Bone Marrow Transplantation , Candida albicans/pathogenicity , Cell Death , Cell Line , Female , Genetic Therapy , Humans , Immunoconjugates/administration & dosage , Mice , Mice, Inbred C57BL , Neoplasms , Tissue Donors
15.
J Am Chem Soc ; 130(31): 10274-81, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18611022

ABSTRACT

Compounds incorporating the triphenylmethyl motif constitute an emerging family of potent anticancer agents. Although several small molecules containing this pharmacophore have now been identified, the mechanism of cell death induction for some of these compounds is unknown. In an effort to define their mechanism of action, and to distinguish subtypes within the group of compounds containing the triphenylmethyl moiety, we have created novel triphenylmethyl-containing small molecules and have evaluated them in a battery of biological assays. Here we show that several phosphonate and phosphonochloridates possessing the triphenylmethyl motif potently induce death of multiple cancer cell lines in culture. Further assays evaluating the ability to cause cell cycle arrest, inhibit tubulin polymerization, dissociate mitochondrial-bound hexokinase in cancer cells, and inhibit calcium-dependent potassium ion channels indicate that triphenylmethyl-containing compounds can be placed into at least four distinct categories, each with a different mechanism of action.


Subject(s)
Antineoplastic Agents/chemistry , Trityl Compounds/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Hexokinase/antagonists & inhibitors , Humans , Organophosphonates , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Structure-Activity Relationship , Trityl Compounds/chemistry , Tubulin/drug effects
16.
Curr Opin Biotechnol ; 18(6): 497-503, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17988854

ABSTRACT

Small molecules that bind to DNA are extremely useful as biochemical tools for the visualization of DNA both in vitro and inside the cell. Additionally, the clinical significance of DNA-binding compounds can hardly be overstated, as many anticancer regimens include a compound that binds to and/or modifies DNA. Although many of the important DNA-binding anticancer drugs were discovered in phenotypic, cell-based screens, in vitro experiments have been developed that enable a precise determination of how a compound interacts with DNA. This review provides a summary of the assays that should be performed when it is suspected that DNA may be a target for a given small molecule. A battery of in vitro assays readily distinguishes between DNA intercalation, DNA groove binding, and the inhibition of topoisomerases. Further cell-based investigations can implicate a direct effect of a compound on DNA within the cell. Together, these assays are powerful tools to determine the mechanism of previously discovered molecules, and will be crucial to the discovery of the next generation of DNA-binding anticancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , DNA/chemistry , Drug Design , Intercalating Agents/chemistry , Binding Sites , Models, Molecular , Molecular Structure , Nucleic Acid Conformation
17.
Bioorg Med Chem Lett ; 18(22): 5888-91, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18710803

ABSTRACT

Triphenylmethylamides (TPMAs) have been previously identified as compounds that arrest cells in the G1-phase of the cell cycle and induce apoptotic death in melanoma cell lines in culture. Here we report the synthesis of a series of TPMA derivatives, allowing the structure-activity relationship of this class of molecules to be established. Several new compounds have been identified that induce death in UACC-62 and SK-MEL-5 human melanoma cell lines, including a compound with enhanced aqueous solubility.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Amides/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , G1 Phase/drug effects , Humans , Molecular Structure , Solubility , Structure-Activity Relationship
18.
Sci Transl Med ; 9(411)2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29021165

ABSTRACT

Targeted genome editing in hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematological diseases. However, the limited efficiency of homology-directed editing in primitive HSPCs constrains the yield of corrected cells and might affect the feasibility and safety of clinical translation. These concerns need to be addressed in stringent preclinical models and overcome by developing more efficient editing methods. We generated a humanized X-linked severe combined immunodeficiency (SCID-X1) mouse model and evaluated the efficacy and safety of hematopoietic reconstitution from limited input of functional HSPCs, establishing thresholds for full correction upon different types of conditioning. Unexpectedly, conditioning before HSPC infusion was required to protect the mice from lymphoma developing when transplanting small numbers of progenitors. We then designed a one-size-fits-all IL2RG (interleukin-2 receptor common γ-chain) gene correction strategy and, using the same reagents suitable for correction of human HSPC, validated the edited human gene in the disease model in vivo, providing evidence of targeted gene editing in mouse HSPCs and demonstrating the functionality of the IL2RG-edited lymphoid progeny. Finally, we optimized editing reagents and protocol for human HSPCs and attained the threshold of IL2RG editing in long-term repopulating cells predicted to safely rescue the disease, using clinically relevant HSPC sources and highly specific zinc finger nucleases or CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9). Overall, our work establishes the rationale and guiding principles for clinical translation of SCID-X1 gene editing and provides a framework for developing gene correction for other diseases.


Subject(s)
Hematopoietic Stem Cells/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Gene Targeting/methods , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Mice , Mice, SCID
19.
Nat Biotechnol ; 34(7): 738-45, 2016 07.
Article in English | MEDLINE | ID: mdl-27272386

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) offers curative therapy for patients with hemoglobinopathies, congenital immunodeficiencies, and other conditions, possibly including AIDS. Autologous HSCT using genetically corrected cells would avoid the risk of graft-versus-host disease (GVHD), but the genotoxicity of conditioning remains a substantial barrier to the development of this approach. Here we report an internalizing immunotoxin targeting the hematopoietic-cell-restricted CD45 receptor that effectively conditions immunocompetent mice. A single dose of the immunotoxin, CD45-saporin (SAP), enabled efficient (>90%) engraftment of donor cells and full correction of a sickle-cell anemia model. In contrast to irradiation, CD45-SAP completely avoided neutropenia and anemia, spared bone marrow and thymic niches, enabling rapid recovery of T and B cells, preserved anti-fungal immunity, and had minimal overall toxicity. This non-genotoxic conditioning method may provide an attractive alternative to current conditioning regimens for HSCT in the treatment of non-malignant blood diseases.


Subject(s)
DNA Damage/immunology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Leukocyte Common Antigens/immunology , Ribosome Inactivating Proteins, Type 1/genetics , Ribosome Inactivating Proteins, Type 1/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA Damage/genetics , Female , Genetic Enhancement/methods , Immunogenetic Phenomena/genetics , Immunotoxins , Mice , Mice, Inbred C57BL , Saporins
20.
Cell Rep ; 13(9): 2027-36, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655912

ABSTRACT

Apoptosis is generally believed to be a process that requires several hours, in contrast to non-programmed forms of cell death that can occur in minutes. Our findings challenge the time-consuming nature of apoptosis as we describe the discovery and characterization of a small molecule, named Raptinal, which initiates intrinsic pathway caspase-dependent apoptosis within minutes in multiple cell lines. Comparison to a mechanistically diverse panel of apoptotic stimuli reveals that Raptinal-induced apoptosis proceeds with unparalleled speed. The rapid phenotype enabled identification of the critical roles of mitochondrial voltage-dependent anion channel function, mitochondrial membrane potential/coupled respiration, and mitochondrial complex I, III, and IV function for apoptosis induction. Use of Raptinal in whole organisms demonstrates its utility for studying apoptosis in vivo for a variety of applications. Overall, rapid inducers of apoptosis are powerful tools that will be used in a variety of settings to generate further insight into the apoptotic machinery.


Subject(s)
Apoptosis , Cyclopentanes/chemistry , Fluorenes/chemistry , Animals , Apoptosis/drug effects , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Complement C8/deficiency , Complement C8/genetics , Cyclopentanes/pharmacokinetics , Cyclopentanes/toxicity , Embryo, Nonmammalian/metabolism , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Fluorenes/pharmacokinetics , Fluorenes/toxicity , Half-Life , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL