Bio


Prof. Agnieszka Czechowicz is a physician-scientist and biotech entrepreneur with 20 years of experience in stem cell biology and translational research. Dr. Czechowicz is a faculty member in Stanford University’s Department of Pediatrics, Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine Division and is also a member of Stanford's Institute for Stem Cell Biology and Regenerative Medicine.

Dr. Czechowicz’s primary clinical interests are in bone marrow failure (including Fanconi anemia and aplastic anemia) and other diverse diseases that necessitate or could benefit from stem cell transplantation. Her primary research interests are in hematopoietic stem cell biology, transplantation, gene-therapy/gene-editing and regenerative medicine. Multiple pre-clinical and clinical therapies are in development based upon her studies.

Dr. Czechowicz’s pioneering work has led to critical developments within the life sciences ecosystem. She was the scientific co-founder of Magenta Therapeutics which amongst other things was developing antibody-drug-conjugates for conditioning for hematopoietic stem cell transplantation, and parallel antibody-based conditioning also based upon her work are additionally being pursued by Jasper Therapeutics and Forty Seven Inc (acquired by Gilead in 2020). Her efforts have also led to important patents that are assigned to Editas Medicine and Decibel Therapeutics. Previously, Dr. Czechowicz spent nearly a decade as a consultant at Third Rock Ventures, where she assisted and started companies including Global Blood Therapeutics, Bluebird Bio, Editas Medicine, Decibel Therapeutics and Magenta Therapeutics. She has also advised Prime Medicines, Beam Therapeutics, and Spotlight Therapeutics.

Dr. Czechowicz completed her Ph.D. work in Developmental Biology at Stanford University with Prof. Irv Weissman. She then completed her residency in Pediatrics at the Boston Children’s Hospital, and subsequently pursued subspecialty training in Pediatric Hematology and Oncology at the Dana Farber Cancer Institute while simultaneously conducting postdoctoral research with Prof. Derrick Rossi in collaboration with Prof. David Scadden. She received her M.D., Ph.D., and Bachelor’s Degree from Stanford University.

Dr. Czechowicz is a strong physician-scientist and advocate of translational research. She has done pioneering work showing that hematopoietic stem cell depletion is a critical component to donor hematopoietic stem cell engraftment. She has also led several clinical trials exploring new cell and gene therapy treatments for Fanconi Anemia. She is passionate about mentoring and training future generations of physicians and scientists, and is very supportive of helping diverse trainees on various traditional and non-traditional career paths. Dr. Czechowicz can best be reached through her administrative assistant Paula Vasquez (paulav22@stanford.edu).

Recent press releases on her efforts can be found here:
https://med.stanford.edu/news/all-news/2019/02/antibody-drug-combo-may-obviate-need-for-tissue-matching.html
http://med.stanford.edu/news/all-news/2019/02/antibody-could-increase-cure-rate-for-blood-immune-disorders.html

Administrative Appointments


  • Member, Center for Definitive and Curative Medicine (CDCM) (2019 - Present)
  • Member, Stanford Institute for Stem Cell Biology and Regenerative Medicine (2018 - Present)
  • Member, Stanford Diabetes Research Center (2018 - Present)

Honors & Awards


  • Lorry I. Lokey Faculty Scholar, Stanford University School of Medicine ((2019-2024))

Professional Education


  • Clinical Fellowship III, Stanford University, Pediatric Hematology/Oncology/Transplant (2017)
  • Clinical Fellowship I-II, Dana Farber Cancer Institute, Pediatric Hematology/Oncology/Transplant (2016)
  • Clinical Residency, Boston Children's Hospital, Pediatrics (2014)

Patents


  • Irving L. Weissman, Agnieszka Czechowicz, Deepta Bhattacharya, Daniel Kraft. "United States Patent Application US12447634 Selective immunodepletion of endogenous stem cell niche for engraftment", Leland Stanford Junior University
  • Alexandra Glucksmann, Deborah Palestrant, Louis Anthony Tartaglia, Jordi Mata-Fink, Agnieszka Czechowicz, Alexis Borisy. "United States Patent Application US14536319 CRISPR-RELATED METHODS AND COMPOSITIONS WITH GOVERNING gRNAS", EDITAS MEDICINE Inc, University of Iowa Research Foundation (UIRF), Massachusetts Institute of Technology, Broad Institute Inc
  • David T. Scadden, Rahul Palchaudhuri, Derrick J. Rossi, Agnieszka Czechowicz. "United States Patent Application US15148837 Compositions and methods for non-myeloablative conditioning", President And Fellows Of Harvard College, The General Hospital Corporation, The Children's Medical Center Corporation
  • Albert Edge, Michael Venuti, Agnieszka Czechowicz. "United States"United States Patent Application US62288958 Expansion and differentiation of inner ear supporting cells and methods of use thereof"", Massachusetts Eye and Ear Infirmary, Decibel Therapeutics

Current Research and Scholarly Interests


Dr. Czechowicz’s research is aimed primarily at understanding how hematopoietic stem cells interact with their microenvironment in order to subsequently modulate these interactions to ultimately improve bone marrow transplantation and unlock biological secrets that further enable regenerative medicine broadly. She is interested in increasing our basic science understanding of these interactions and also developing new novel therapies that stem from this work to expand treatment options for a wide variety of pediatric and adult diseases. Her group is primarily focused on studying the cell surface receptors on hematopoietic stem/progenitor cells and bone marrow stromal cells, and is actively learning how manipulating these can alter cell state and cell fate. Her group is using cells and serum from both mice and primary specimens from healthy and diseased patients for these studies and using a variety of exciting new tools and methods to unlock future discoveries. There are many exciting opportunities that stem from her work across a variety of disease states ranging from rare genetic diseases, autoimmune diseases, solid organ transplantation, microbiome and cancer. While her group is primarily focused on blood and immune diseases, the expanded potential of this work is much broader and can be applied to other organ systems as well and she is very eager to develop collaborations across disease areas.

Dr. Czechowicz has also been part of the initial founding team of several companies including Global Blood Therapeutics, Editas Medicine, Decibel Therapeutics and Magenta Therapeutics and advises multiple other transformative companies. As a true physician scientist, she has done pioneering work showing that hematopoietic stem cell depletion is a critical component to donor hematopoietic stem cell engraftment, and multiple pre-clinical and clinical therapies are in development based upon her studies. Stanford has had several clinical trials derived from Dr. Czechowicz’s research.

Research Interests: Hematopoietic Stem Cells (HSC), Hematopoietic Stem Cell Transplantation (HSCT), Monoclonal Antibodies, Immunotoxins, Cell Cycle, Cell Fate, Cell Membrane, Cell Surface Antigens, Microenvironment, Stem Cell Niche, Cell Proliferation, Stem Cell Quiescence, DNA Damage, DNA Repair, Rare Genetic Diseases, Bone Marrow Failure, Aplastic Anemia, Genomics, Fanconi Anemia (FA), Immunodeficiency (SCID), Gastrointestinal Stromal Tumors (GIST), Rhabdomyosarcoma, Neuroblastoma, Myelodysplastic Syndrome (MDS), Acute Myeloid Leukemia (AML), Graft vs Host Disease (GVHD), Immune Tolerance, Histocompatibility Testing, Immunologic Deficiency Syndromes, Hemoglobinopathies, Transplantation Conditioning, Immune Tolerance, Gene Therapy, Gene-Editing, Base-Editing, Cytokines, Cytokine Receptors, Serum, Clinical Trials, Autoimmune diseases, Multiple Sclerosis, Microbiome, Cancer, Cell Therapy, Allogenic Bone Marrow Transplantation (BMT), Metabolic Diseases, Hurler Syndrome

For more information, please visit our lab webpage: http://med.stanford.edu/czechowiczlab.html

Clinical Trials


  • A Clinical Trial to Evaluate the Safety of RP-L102 in Pediatric Subjects With Fanconi Anemia Subtype A Recruiting

    The objective of this study is to assess the therapeutic safety and preliminary efficacy of a hematopoietic cell-based gene therapy consisting of autologous CD34+ enriched cells transduced with a lentiviral vector carrying the FANCA gene in subjects with Fanconi anemia subtype A (FA-A).

    View full details

  • Depleted Donor Stem Cell Transplant in Children and Adults With Fanconi Anemia After Being Conditioned With a Regimen Containing Briquilimab Recruiting

    The objective of this clinical trial is to develop a cell therapy for Fanconi Anemia which enables enhanced donor hematopoietic and immune reconstitution with decreased toxicity by transplanting depleted stem cells from a donor after using an experimental antibody treatment called JSP-191 as a part of conditioning. This experimental treatment will hopefully cause fewer side effects than chemotherapy (the current standard of care method). Participants will be administered the conditioning regimen, are assessed until they receive the depleted stem cell infusion, and will be followed for up to 2 years after the cell infusion.

    View full details

  • Gene Therapy for Fanconi Anemia, Complementation Group A Recruiting

    The objective of this study is to assess the therapeutic efficacy of a hematopoietic cell-based gene therapy for patients with Fanconi anemia, subtype A (FA-A). Hematopoietic stem cells from mobilized peripheral blood of patients with FA-A will be transduced ex vivo (outside the body) with a lentiviral vector carrying the FANCA gene. After transduction, the corrected stem cells will be infused intravenously back to the patient with the goal of preventing bone marrow failure.

    View full details

2023-24 Courses


Stanford Advisees


All Publications


  • Anti-CD117 antibody depletes normal and myelodysplastic syndrome human hematopoietic stem cells in xenografted mice. Blood Pang, W. W., Czechowicz, A., Logan, A. C., Bhardwaj, R., Poyser, J., Park, C. Y., Weissman, I. L., Shizuru, J. A. 2019

    Abstract

    The myelodysplastic syndromes (MDS) represent a group of clonal disorders that result in ineffective hematopoiesis and are associated with an increased risk of transformation into acute leukemia. MDS arises from hematopoietic stem cells (HSCs); therefore, successful elimination of MDS HSCs is an important part of any curative therapy. However, current treatment options, including allogeneic hematopoietic cell transplantation (HCT), often fail to ablate disease-initiating MDS HSCs, and thus have low curative potential and high relapse rates. Here, we demonstrate that human HSCs can be targeted and eliminated by monoclonal antibodies (mAbs) that bind cell surface CD117 (c-Kit). We show that an anti-human CD117 mAb, SR-1, inhibits normal cord blood and bone marrow HSCs in vitro. Further, SR-1 and clinical-grade humanized anti-human CD117 mAb, AMG 191, deplete normal and MDS HSCs in vivo in xenograft mouse models. Anti-CD117 mAbs also facilitate the engraftment of normal donor human HSCs in MDS xenograft mouse models, restoring normal human hematopoiesis and eradicating aggressive pathologic MDS cells. This study is the first to demonstrate that anti-human CD117 mAbs have potential as novel therapeutics to eradicate MDS HSCs and augment the curative effect of allogeneic HCT for this disease. Moreover, we establish the foundation for use of these antibody agents not only in the treatment of MDS but for the multitude of other HSC-driven blood and immune disorders for which transplant can be disease-altering.

    View details for PubMedID 30745302

  • Hematopoietic chimerism and donor-specific skin allograft tolerance after non-genotoxic CD117 antibody-drug-conjugate conditioning in MHC-mismatched allotransplantation. Nature communications Li, Z., Czechowicz, A., Scheck, A., Rossi, D. J., Murphy, P. M. 2019; 10 (1): 616

    Abstract

    Hematopoietic chimerism after allogeneic bone marrow transplantation may establish a state of donorantigen-specific tolerance. However, current allotransplantation protocols involve genotoxic conditioning which has harmful side-effects and predisposes to infection and cancer. Here we describe a non-genotoxic conditioning protocol for fully MHC-mismatched bone marrow allotransplantation in mice involving transient immunosuppression and selective depletion of recipient hematopoietic stem cells with a CD117-antibody-drug-conjugate (ADC). This protocol resulted in multilineage, high level (up to 50%), durable, donor-derived hematopoietic chimerism after transplantation of 20 million total bone marrow cells, compared with ≤2.1% hematopoietic chimerism from 50 million total bone marrow cells without conditioning. Moreover, long-term survival of bone marrow donor-type but not third partyskin allografts is achieved in CD117-ADC-conditioned chimeric mice without chronic immunosuppression. The only observed adverse event is transient elevation of liver enzymes in the first week after conditioning. These results provide proof-of-principle for CD117-ADC as a non-genotoxic, highly-targeted conditioning agent in allotransplantation and tolerance protocols.

    View details for PubMedID 30728353

  • Selective hematopoietic stem cell ablation using CD117-antibody-drug-conjugates enables safe and effective transplantation with immunity preservation. Nature communications Czechowicz, A., Palchaudhuri, R., Scheck, A., Hu, Y., Hoggatt, J., Saez, B., Pang, W. W., Mansour, M. K., Tate, T. A., Chan, Y. Y., Walck, E., Wernig, G., Shizuru, J. A., Winau, F., Scadden, D. T., Rossi, D. J. 2019; 10 (1): 617

    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.

    View details for PubMedID 30728354

  • Clonal-level lineage commitment pathways of hematopoietic stem cells in vivo. Proceedings of the National Academy of Sciences of the United States of America Lu, R., Czechowicz, A., Seita, J., Jiang, D., Weissman, I. L. 2019

    Abstract

    While the aggregate differentiation of the hematopoietic stem cell (HSC) population has been extensively studied, little is known about the lineage commitment process of individual HSC clones. Here, we provide lineage commitment maps of HSC clones under homeostasis and after perturbations of the endogenous hematopoietic system. Under homeostasis, all donor-derived HSC clones regenerate blood homogeneously throughout all measured stages and lineages of hematopoiesis. In contrast, after the hematopoietic system has been perturbed by irradiation or by an antagonistic anti-ckit antibody, only a small fraction of donor-derived HSC clones differentiate. Some of these clones dominantly expand and exhibit lineage bias. We identified the cellular origins of clonal dominance and lineage bias and uncovered the lineage commitment pathways that lead HSC clones to different levels of self-renewal and blood production under various transplantation conditions. This study reveals surprising alterations in HSC fate decisions directed by conditioning and identifies the key hematopoiesis stages that may be manipulated to control blood production and balance.

    View details for PubMedID 30622181

  • Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches SCIENCE Czechowicz, A., Kraft, D., Weissman, I. L., Bhattacharya, D. 2007; 318 (5854): 1296-1299

    Abstract

    Upon intravenous transplantation, hematopoietic stem cells (HSCs) can home to specialized niches, yet most HSCs fail to engraft unless recipients are subjected to toxic preconditioning. We provide evidence that, aside from immune barriers, donor HSC engraftment is restricted by occupancy of appropriate niches by host HSCs. Administration of ACK2, an antibody that blocks c-kit function, led to the transient removal of >98% of endogenous HSCs in immunodeficient mice. Subsequent transplantation of these mice with donor HSCs led to chimerism levels of up to 90%. Extrapolation of these methods to humans may enable mild but effective conditioning regimens for transplantation.

    View details for DOI 10.1126/science.1149726

    View details for Web of Science ID 000251086600042

    View details for PubMedID 18033883

    View details for PubMedCentralID PMC2527021

  • Radiation and Busulfan-Free Hematopoietic Stem Cell Transplantation Using Briquilimab (JSP191) Anti-CD117 Antibody-Conditioning, Transient Immunosuppression and TCR α β + T-Cell/CD19+B-Cell Depleted Haploidentical Grafts in Patients with Fanconi Anemia Agarwal, R., Bertaina, A., Soco, C., Saini, G., Kunte, N., Hiroshima, L., Chan, Y., Willner, H., Krampf, M. L., Nofal, R., Barbarito, G., Sen, S., Felber, M., Van Hentenryck, M., Walck, E., Scheck, A., Thongthip, S., Logan, A. C., Dougall, K., Bouge, A., Boelens, J., Long-Boyle, J. R., Weissman, I. L., Shizuru, J., Pang, W. W., Weinberg, K. I., Parkman, R., Roncarolo, M., Porteus, M., Czechowicz, A. AMER SOC HEMATOLOGY. 2023
  • LENTIVIRAL-MEDIATED GENE THERAPY FOR PATIENTS WITH FANCONI ANEMIA [GROUP A]: UPDATED RESULTS FROM GLOBAL RP-L102 CLINICAL TRIALS Sevilla, J., Booth, C., Czechowicz, A., Agarwal, R., Zubicaray, J., Rio, P., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Eide, C., Wagner, J., Rao, G., Thrasher, A., Schwartz, J., Roncarolo, M., Bueren, J. SPRINGERNATURE. 2023: 276-277
  • LENTIVIRAL-MEDIATED GENE THERAPY FOR FANCONI ANEMIA [GROUP A]: RESULTS FROM RP-L102 CLINICAL TRIALS Czechowicz, A., Sevilla, J., Booth, C., Agarwal, R., Zubicaray, J., Rio, P., Navarro, S., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Wagner, J., Eide, C., Rao, G., Thrasher, A., Schwartz, J., Roncarolo, M., Bueren, J. WILEY. 2023: S136-S137
  • Lentiviral-Mediated Gene Therapy for Fanconi Anemia [Group A]: Results from Global RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Booth, C., Navarro, S., Agarwal, R., Zubicaray, J., Rio, P., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Eide, C., Wagner, J. E., Rao, G., Thrasher, A. J., Schwartz, J., Roncarolo, M., Bueren, J. A. CELL PRESS. 2023: 118
  • Lentiviral-mediated Gene Therapy for Patients with Fanconi Anemia [Group A]: Updated Results from Global RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Booth, C., Agarwal, R., Zubicaray, J., Rio, P., Navarro, S., Chetty, K., O'Toole, G., Xu-Bayford, J., Ancliff, P., Sebastian, E., Choi, G., Zeini, M., Nicoletti, E., Wagner, J. E., Rao, G. R., Thrasher, A. J., Schwartz, J. D., Roncarolo, M., Bueren, J. A. AMER SOC HEMATOLOGY. 2022: 10646-10647
  • Hematopoietic and Immunological Assessment in Fanconi Anemia after Ex Vivo Lentiviral FANCA Gene Therapy with RP-L102 Nofal, R., Chan, Y., Sen, S., Figueroa, U., Willner, H., Felber, M., Krampf, M., Thongthip, S., Choi, G., Nicoletti, E., Schwartz, J. D., Weinberg, K., Rodriguez, A., Agarwal, R., Roncarolo, M., Czechowicz, A. AMER SOC HEMATOLOGY. 2022: 7772-7773
  • Non-Genotoxic Restoration of the Hematolymphoid System in Fanconi Anemia Mice through Antibody-Mediated Hematopoietic Stem Cell Transplantation Chan, Y., Ho, P., Swartzrock, L., Rayburn, M., Nofal, R., Thongthip, S., Weinberg, K. I., Czechowicz, A. AMER SOC HEMATOLOGY. 2022: 7342-7344
  • Non-Genotoxic Restoration of the Hematolymphoid System in Fanconi Anemia. Transplantation and cellular therapy Chan, Y. Y., Ho, P. Y., Swartzrock, L., Rayburn, M., Nofal, R., Thongthip, S., Weinberg, K. I., Czechowicz, A. 2022

    Abstract

    BACKGROUND: Hematopoietic stem cell transplantation (HSCT) is a curative treatment for patients with many different blood and immune diseases; however, current treatment regimens contain non-specific chemotherapy and/or irradiation conditioning which have both short-term and long-term toxicities. Use of such agents may be particularly harmful for Fanconi Anemia (FA) patients who have genetic mutations resulting in deficiencies in DNA repair leading to an increased sensitivity to genotoxic agents.OBJECTIVE: Monoclonal antibody (mAb)-based conditioning has been proposed as an alternative conditioning strategy for HSCT that minimizes toxicities by eliminating collateral tissue damage. Given the high need for improved treatments for FA patients, we aimed to evaluate the efficacy of different alphaCD117 mAb agents and immunosuppression on HSC depletion and explored their ability to safely establish therapeutic donor hematopoiesis post HSCT in FA disease models.STUDY DESIGN: We evaluated the efficacy of different concentration of alphaCD117 mAb in vitro and in vivo for HSC growth and depletion. To further assess the efficacy of mAb-based conditioning, Fancd2-/- animals were treated with alphaCD117 mAb and combination agents with alphaCD47 mAb and antibody-drug-conjugates (ADC) for syngeneic HSCT (n=3-5). Immunosuppression alphaCD4 mAb was added to all in vivo experiments due to slight mismatched background between the donor grafts and recipients. Immunosuppressant cocktails were also given to Fancd2-/- animals to evaluate the efficacy of mAb-based conditioning in haploidentical setting (n=5). All statistics was calculated using unpaired t-test.RESULTS: We found that antagonistic alphaCD117 mAbs alone do not deplete host HSCs nor enhance HSCT effectively in FA mouse models; however, the potency of alphaCD117 mAbs can be safely augmented 1) in combination with alphaCD47 mAbs and 2) with antibody-drug-conjugates (ADCs) which both lead to profound HSC depletion and establish long-term donor engraftment post syngeneic HSCT. This is the first time these approaches have been tested in parallel in any disease setting with the highest donor engraftment observed post CD117-ADC conditioning. Interestingly, our data also suggests that HSC-targeted conditioning is surprisingly not necessary in FA HSCT as high donor HSC engraftment was observed with mAb-based immune suppression alone with immunologically matched and mismatched haploidentical grafts.CONCLUSIONS: These results demonstrate the safety and efficacy of several different non-genotoxic mAb-based conditioning strategies in the FA setting. In addition, they show that if sufficient immunosuppression is given to obtain initial donor HSC engraftment, resulting turnover of a majority of the hematolymphoid system can occur likely due to the survival advantage of WT HSCs over FA HSCs. Such non-toxic all antibody-based conditioning strategy could be transformative for FA patients and those with other hematolymphoid diseases.

    View details for DOI 10.1016/j.jtct.2022.08.015

    View details for PubMedID 35995393

  • Gene Therapy for Fanconi Anemia [Group A]: Interim Results of RP-L102 Clinical Trials Czechowicz, A., Sevilla, J., Agarwal, R., Booth, C., Zubicaray, J., Rio, P., Navarro, S., Ancliff, P., Sebastian, E., Beard, B. C., Law, K. M., Choi, G., Zeini, M., Duran-Persson, C., Nicoletti, E., Wagner, J. E., Rao, G. R., Thrasher, A. J., Schwartz, J. D., Bueren, J. A., Roncarolo, M. AMER SOC HEMATOLOGY. 2021
  • Peptide nucleic acid-dependent artifact can lead to false-positive triplex gene editing signals. Proceedings of the National Academy of Sciences of the United States of America Ho, P. Y., Zhang, Z., Hayes, M. E., Curd, A., Dib, C., Rayburn, M., Tam, S. N., Srivastava, T., Hriniak, B., Li, X., Leonard, S., Wang, L., Tarighat, S., Sim, D. S., Fiandaca, M., Coull, J. M., Ebens, A., Fordyce, M., Czechowicz, A. 2021; 118 (45)

    Abstract

    Triplex gene editing relies on binding a stable peptide nucleic acid (PNA) sequence to a chromosomal target, which alters the helical structure of DNA to stimulate site-specific recombination with a single-strand DNA (ssDNA) donor template and elicits gene correction. Here, we assessed whether the codelivery of PNA and donor template encapsulated in Poly Lactic-co-Glycolic Acid (PLGA)-based nanoparticles can correct sickle cell disease and x-linked severe combined immunodeficiency. However, through this process we have identified a false-positive PCR artifact due to the intrinsic capability of PNAs to aggregate with ssDNA donor templates. Here, we show that the combination of PNA and donor templates but not either agent alone results in different degrees of aggregation that result in varying but highly reproducible levels of false-positive signal. We have identified this phenomenon in vitro and confirmed that the PNA sequences producing the highest supposed correction in vitro are not active in vivo in both disease models, which highlights the importance of interrogating and eliminating carryover of ssDNA donor templates in assessing various gene editing technologies such as PNA-mediated gene editing.

    View details for DOI 10.1073/pnas.2109175118

    View details for PubMedID 34732575

  • Safe and Effective In Vivo Targeting and Gene Editing in Hematopoietic Stem Cells: Strategies for Accelerating Development National Institutes of Health/Bill & Melinda Gates Foundation Expert Scientific Roundtable Webinar Meeting. Human gene therapy Cannon, P., Asokan, A., Czechowicz, A., Hammond, P., Kohn, D. B., Lieber, A., Malik, P., Marks, P., Porteus, M., Verhoeyen, E., Weissman, D., Weissman, I., Kiem, H. 2021

    Abstract

    Introduction On May 11, 2020, the National Institutes of Health (NIH) and the Bill & Melinda Gates Foundation (Gates Foundation) held an exploratory expert scientific roundtable to inform an NIH-Gates Foundation collaboration on the development of scalable, sustainable, and accessible HIV and sickle cell disease (SCD) therapies based on in vivo gene editing of hematopoietic stem cells (HSC). A particular emphasis was on how such therapies could be developed for low-resource settings in sub-Saharan Africa. Paula Cannon, Ph.D., of the University of Southern California and Hans-Peter Kiem, M.D., Ph.D., of the Fred Hutchinson Cancer Research Center served as roundtable co-chairs. Welcoming remarks were provided by the leadership of NIH, NHLBI, and BMGF, who cited the importance of assessing the state of the science and charting a path toward finding safe, effective, and durable gene-based therapies for HIV and sickle cell disease. These remarks were followed by three sessions in which participants heard presentations on and discussed the therapeutic potential of modified HSCs, leveraging HSC biology and differentiation, and in vivo HSC targeting approaches. This roundtable serves as the beginning of an ongoing discussion among NIH, the Gates Foundation, research and patient communities, and the public at large. As this collaboration progresses, these communities will be engaged as we collectively navigate the complex scientific and ethical issues surrounding in vivo HSC targeting and editing. Summarized excerpts from each of the presentations are below, reflecting the individual views and perspectives of each presenter.

    View details for DOI 10.1089/hum.2020.263

    View details for PubMedID 33427035

  • A beta T-Cell/CD19 B-Cell Depleted Haploidentical Stem Cell Transplantation: A New Platform for Curing Rare and Monogenic Disorders Bertaina, A., Bacchetta, R., Lewis, D. B., Grimm, P. C., Shah, A. J., Agarwal, R., Concepcion, W., Czechowicz, A., Bhatia, N., Lahiri, P., Weinberg, K. I., Parkman, R., Porteus, M., Roncarolo, M. ELSEVIER SCIENCE INC. 2020: S288
  • Changing the Natural History of Fanconi Anemia Complementation Group-A with Gene Therapy: Early Results of US Phase I Study of Lentiviral-Mediated Ex-Vivo FANCA Gene Insertion in Human Stem and Progenitor Cells Czechowicz, A., Rio, P., Bueren, J. E., Beard, B., Nicoletti, E., Schwartz, J., Soni, S. ELSEVIER SCIENCE INC. 2020: S39–S40
  • Nongenotoxic antibody-drug conjugate conditioning enables safe and effective platelet gene therapy of hemophilia A mice. Blood advances Gao, C., Schroeder, J. A., Xue, F., Jing, W., Cai, Y., Scheck, A., Subramaniam, S., Rao, S., Weiler, H., Czechowicz, A., Shi, Q. 2019; 3 (18): 2700–2711

    Abstract

    Gene therapy offers the potential to cure hemophilia A (HA). We have shown that hematopoietic stem cell (HSC)-based platelet-specific factor VIII (FVIII) (2bF8) gene therapy can produce therapeutic protein and induce antigen-specific immune tolerance in HA mice, even in the presence of inhibitory antibodies. For HSC-based gene therapy, traditional preconditioning using cytotoxic chemotherapy or total body irradiation (TBI) has been required. The potential toxicity associated with TBI or chemotherapy is a deterrent that may prevent patients with HA, a nonmalignant disease, from agreeing to such a protocol. Here, we describe targeted nongenotoxic preconditioning for 2bF8 gene therapy utilizing a hematopoietic cell-specific antibody-drug conjugate (ADC), which consists of saporin conjugated to CD45.2- and CD117-targeting antibodies. We found that a combination of CD45.2- and CD117-targeting ADC preconditioning was effective for engrafting 2bF8-transduced HSCs and was favorable for platelet lineage reconstitution. Two thirds of HA mice that received 2bF8 lentivirus-transduced HSCs under (CD45.2+CD117)-targeting ADC conditioning maintained sustained therapeutic levels of platelet FVIII expression. When CD8-targeting ADC was supplemented, chimerism and platelet FVIII expression were significantly increased, with long-term sustained platelet FVIII expression in all primary and secondary recipients. Importantly, immune tolerance was induced and hemostasis was restored in a tail-bleeding test, and joint bleeding also was effectively prevented in a needle-induced knee joint injury model in HA mice after 2bF8 gene therapy. In summary, we show for the first time efficient engraftment of gene-modified HSCs without genotoxic conditioning. The combined cocktail ADC-mediated hematopoietic cell-targeted nongenotoxic preconditioning that we developed is highly effective and favorable for platelet-specific gene therapy in HA mice.

    View details for DOI 10.1182/bloodadvances.2019000516

    View details for PubMedID 31515232

  • Anti-CD117 antibody depletes normal and myelodysplastic syndrome human hematopoietic stem cells in xenografted mice BLOOD Pang, W. W., Czechowicz, A., Logan, A. C., Bhardwaj, R., Poyser, J., Park, C. Y., Weissman, I. L., Shizuru, J. A. 2019; 133 (19): 2069–78
  • Anti-human CD117 antibody-mediated bone marrow niche clearance in nonhuman primates and humanized NSG mice BLOOD Kwon, H., Logan, A. C., Chhabra, A., Pang, W. W., Czechowicz, A., Tate, K., Le, A., Poyser, J., Hollis, R., Kelly, B. V., Kohn, D. B., Weissman, I. L., Prohaska, S. S., Shizuru, J. A. 2019; 133 (19): 2104–8
  • Anti-human CD117 antibody-mediated bone marrow niche clearance in non-human primates and humanized NSG mice. Blood Kwon, H., Logan, A. C., Chhabra, A., Pang, W. W., Czechowicz, A., Tate, K., Le, A., Poyser, J., Hollis, R., Kelly, B. V., Kohn, D. B., Weissman, I. L., Prohaska, S. S., Shizuru, J. A. 2019

    View details for PubMedID 30617195

  • The MarrowMiner: A Novel Minimally Invasive and Effective Device for the Harvest of Bone Marrow. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation Kraft, D. n., Walck, E. n., Carrasco, A. n., Crocker, M. n., Song, L. n., Long, M. n., Mosse, M. n., Nadeem, B. n., Imanbyev, G. n., Czechowicz, A. n., McCullough, M. n. 2019

    Abstract

    Bone marrow (BM) is a rich source of hematopoietic stem cells (HSC), mesenchymal stem cells (MSC) and other important stem/progenitor cells. It is the traditional source of cells utilized in hematopoietic cell transplantation (HCT), which is a proven curative treatment for many blood and immune diseases. BM-derived cells have also been shown to have other diverse clinical uses and are increasingly being utilized in orthopedic, regenerative medicine, and gene therapy applications. Traditional methods for harvesting BM are crude, tedious, time-consuming and expensive; requiring multiple bone punctures under general anesthesia with serial small volume aspirates often diluted with peripheral blood. The MarrowMiner (MM) is a novel device designed for rapid and minimally invasive BM harvest. Here we show the safety and efficacy of the MM in both preclinical and clinical settings. In a large-animal porcine model, the MM enabled effective BM collection with similar total nucleated cell collection and increased colony formation (CFU-F) compared to standard methods. The MM was subsequently evaluated in a clinical study showing effective and complication-free anterior and posterior BM collection of 20 subjects under only local anesthesia or light sedation. Increased total nucleated and mononucleated cell collection was achieved with the MM compared to standard methods in the same subjects. Importantly, stem cell content was high with trends towards increased HSC, MSC and endothelial progenitor cells with similar T-cell content. Given the MarrowMiner is a novel, FDA-approved device, enabling safe, effective and minimally invasive harvest of BM we anticipate rapid adoption for various applications.

    View details for DOI 10.1016/j.bbmt.2019.08.027

    View details for PubMedID 31491487

  • Selective HSC-Ablation Using CD117 Antibody-Drug-Conjugates Enables Safe and Effective Murine and Human Hematopoietic Stem Cell Transplantation Czechowicz, A., Palchaudhuri, R., Scheck, A., Hu, Y., Hoggatt, J., Saez, B., Pang, W. W., Mansour, M. K., Shizuru, J. A., Winau, F., Scadden, D. T., Rossi, D. J. CELL PRESS. 2018: 23–24
  • Safe and Effective Platelet-Targeted Gene Therapy of Hemophilia A Enabled Using Non-Genotoxic, Antibody-Drug-Conjugate Conditioning Gao, C., Schroeder, J., Czechowicz, A., Shi, Q. CELL PRESS. 2018: 25
  • Immune Sparing Conditioning for BMT/HSCT Using Anti-Ckit Immunotoxins Czechowicz, A., Palchaudhuri, R., Scheck, A., Hu, Y., Winau, F., Hoggatt, J., Saez, B., Mansour, M. K., Sykes, D., Scadden, D., Rossi, D. ELSEVIER SCIENCE INC. 2018: S60–S61
  • Anti-Human CD117 Antibodies Mediate Clearance of Myelodysplastic Syndrome Hematopoietic Stem Cells and Facilitate Establishment of Normal Hematopoiesis in Transplantation Pang, W. W., Czechowicz, A., Poyser, J., Park, C. Y., Weissman, I. L., Shizuru, J. A. ELSEVIER SCIENCE INC. 2018: S230–S231
  • Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin NATURE BIOTECHNOLOGY Palchaudhuri, R., Saez, B., Hoggatt, J., Schajnovitz, A., Sykes, D. B., Tate, T. A., Czechowicz, A., Kfoury, Y., Ruchika, F. N., Rossi, D. J., Verdine, G. L., Mansour, M. K., Scadden, D. T. 2016; 34 (7): 738-?

    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.

    View details for DOI 10.1038/nbt.3584

    View details for Web of Science ID 000381335400028

    View details for PubMedID 27272386

    View details for PubMedCentralID PMC5179034

  • A trial of plerixafor adjunctive therapy in allogeneic hematopoietic cell transplantation with minimal conditioning for severe combined immunodeficiency PEDIATRIC TRANSPLANTATION Dvorak, C. C., Horn, B. N., Puck, J. M., Czechowicz, A., Shizuru, J. A., Ko, R. M., Cowan, M. J. 2014; 18 (6): 602-608

    Abstract

    For infants with SCID, the ideal conditioning regimen before allogeneic HCT would omit cytotoxic chemotherapy to minimize short- and long-term complications. We performed a prospective pilot trial with G-CSF plus plerixafor given to the host to mobilize HSC from their niches. We enrolled six patients who received CD34-selected haploidentical cells and one who received T-replete matched unrelated BM. All patients receiving G-CSF and plerixafor had generally poor CD34(+) cell and Lin(-) CD34(+) CD38(-) CD90(+) CD45RA(-) HSC mobilization, and developed donor T cells, but no donor myeloid or B-cell engraftment. Although well tolerated, G-CSF plus plerixafor alone failed to overcome physical barriers to donor engraftment.

    View details for DOI 10.1111/petr.12309

    View details for Web of Science ID 000340530800017

    View details for PubMedCentralID PMC5413354

  • A trial of alemtuzumab adjunctive therapy in allogeneic hematopoietic cell transplantation with minimal conditioning for severe combined immunodeficiency PEDIATRIC TRANSPLANTATION Dvorak, C. C., Horn, B. N., Puck, J. M., Adams, S., Veys, P., Czechowicz, A., Cowan, M. J. 2014; 18 (6): 609-616

    Abstract

    For infants with SCID the ideal conditioning regimen before allogeneic HCT would omit cytotoxic chemotherapy to minimize short- and long-term complications. We performed a prospective pilot trial with alemtuzumab monotherapy to overcome NK-cell mediated immunologic barriers to engraftment. We enrolled four patients who received CD34-selected haploidentical cells, two of whom failed to engraft donor T cells. The two patients who engrafted had delayed T-cell reconstitution, despite rapid clearance of circulating alemtuzumab. Although well-tolerated, alemtuzumab failed to overcome immunologic barriers to donor engraftment. Furthermore, alemtuzumab may slow T-cell development in patients with SCID in the setting of a T-cell depleted graft.

    View details for DOI 10.1111/petr.12310

    View details for Web of Science ID 000340530800018

    View details for PubMedID 24977928

    View details for PubMedCentralID PMC4134761

  • In utero depletion of fetal hematopoietic stem cells improves engraftment after neonatal transplantation in mice. Blood Derderian, S. C., Togarrati, P. P., King, C., Moradi, P. W., Reynaud, D., Czechowicz, A., Weissman, I. L., MacKenzie, T. C. 2014; 124 (6): 973-980

    Abstract

    Although in utero hematopoietic cell transplantation is a promising strategy to treat congenital hematopoietic disorders, levels of engraftment have not been therapeutic for diseases in which donor cells have no survival advantage. We used an antibody against the murine c-Kit receptor (ACK2) to deplete fetal host hematopoietic stem cells (HSCs) and increase space within the hematopoietic niche for donor cell engraftment. Fetal mice were injected with ACK2 on embryonic days 13.5 to 14.5 and surviving pups were transplanted with congenic hematopoietic cells on day of life 1. Low-dose ACK2 treatment effectively depleted HSCs within the bone marrow with minimal toxicity and the antibody was cleared from the serum before the neonatal transplantation. Chimerism levels were significantly higher in treated pups than in controls; both myeloid and lymphoid cell chimerism increased because of higher engraftment of HSCs in the bone marrow. To test the strategy of repeated HSC depletion and transplantation, some mice were treated with ACK2 postnatally, but the increase in engraftment was lower than that seen with prenatal treatment. We demonstrate a successful fetal conditioning strategy associated with minimal toxicity. Such strategies could be used to achieve clinically relevant levels of engraftment to treat congenital stem cell disorders.

    View details for DOI 10.1182/blood-2014-02-550327

    View details for PubMedID 24879814

  • Anti-KIT monoclonal antibody inhibits imatinib-resistant gastrointestinal stromal tumor growth PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Edris, B., Willingham, S. B., Weiskopf, K., Volkmer, A. K., Volkmer, J., Muehlenberg, T., Montgomery, K. D., Contreras-Trujillo, H., Czechowicz, A., Fletcher, J. A., West, R. B., Weissman, I. L., van de Rijn, M. 2013; 110 (9): 3501-3506

    Abstract

    Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract and arises from the interstitial cells of Cajal. It is characterized by expression of the receptor tyrosine kinase CD117 (KIT). In 70-80% of GIST cases, oncogenic mutations in KIT are present, leading to constitutive activation of the receptor, which drives the proliferation of these tumors. Treatment of GIST with imatinib, a small-molecule tyrosine kinase inhibitor, inhibits KIT-mediated signaling and initially results in disease control in 70-85% of patients with KIT-positive GIST. However, the vast majority of patients eventually develop resistance to imatinib treatment, leading to disease progression and posing a significant challenge in the clinical management of these tumors. Here, we show that an anti-KIT monoclonal antibody (mAb), SR1, is able to slow the growth of three human GIST cell lines in vitro. Importantly, these reductions in cell growth were equivalent between imatinib-resistant and imatinib-sensitive GIST cell lines. Treatment of GIST cell lines with SR1 reduces cell-surface KIT expression, suggesting that mAb-induced KIT down-regulation may be a mechanism by which SR1 inhibits GIST growth. Furthermore, we also show that SR1 treatment enhances phagocytosis of GIST cells by macrophages, indicating that treatment with SR1 may enhance immune cell-mediated tumor clearance. Finally, using two xenotransplantation models of imatinib-sensitive and imatinib-resistant GIST, we demonstrate that SR1 is able to strongly inhibit tumor growth in vivo. These results suggest that treatment with mAbs targeting KIT may represent an alternative, or complementary, approach for treating GIST.

    View details for DOI 10.1073/pnas.1222893110

    View details for PubMedID 23382202

  • Anti-CD117 (c-Kit) Monoclonal Antibodies Deplete Human Hematopoietic Stem Cells and Facilitate Their Replacement in Humanized NOD/SCID/IL2R gamma(-/-) Mice: A Non-Toxic Conditioning Regimen for Allotransplantation Logan, A. C., Czechowicz, A., Kelley, B. V., Thway, T. M., Magana, I., Krampf, M. R., Poyser, J., Hollis, R. P., Kohn, D. B., Weissman, I. L., Shizuru, J. A. AMER SOC HEMATOLOGY. 2012
  • Clonal Level Lineage Commitment of Mouse Hematopoietic Stem Cells in Vivo 54th Annual Meeting and Exposition of the American-Society-of-Hematology (ASH) Lu, R., Czechowicz, A., Seita, J., Weissman, I. L. AMER SOC HEMATOLOGY. 2012
  • Transplantation Conditioning Regimens Induce Different Hematopoietic Stem Cell Differentiation in Mice At the Clonal Level 53rd Annual Meeting and Exposition of the American-Society-of-Hematology (ASH) Lu, R., Czechowicz, A., Weissman, I. L. AMER SOC HEMATOLOGY. 2011: 67–67
  • Purified Hematopoietic Stem Cell Transplantation: The Next Generation of Blood and Immune Replacement HEMATOLOGY-ONCOLOGY CLINICS OF NORTH AMERICA Czechowicz, A., Weissman, I. L. 2011; 25 (1): 75-?

    Abstract

    Replacement of disease-causing stem cells with healthy ones has been achieved clinically via hematopoietic cell transplantation (HCT) for the last 40 years, as a treatment modality for a variety of cancers and immunodeficiencies with moderate, but increasing, success. This procedure has traditionally included transplantation of mixed hematopoietic populations that include hematopoietic stem cells (HSC) and other cells, such as T cells. This article explores and delineates the potential expansion of this technique to treat a variety of inherited diseases of immune function, the current barriers in HCT and pure HSC transplantation, and the up-and-coming strategies to combat these obstacles.

    View details for DOI 10.1016/j.hoc.2010.11.006

    View details for Web of Science ID 000287333600007

    View details for PubMedID 21236391

  • Targeted Clearance of Human Hematopoietic Stem Cell Niches Via Inhibition of SCF Signaling Using Monoclonal Antibody SR-1 52nd Annual Meeting and Exposition of the American-Society-of-Hematology (ASH) Czechowicz, A., Bhardwaj, R., Park, C. Y., Weissman, I. L. AMER SOC HEMATOLOGY. 2010: 39–40
  • Inhibition of Mac-1 (CD11b/CD18) enhances tumor response to radiation by reducing myeloid cell recruitment PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Ahn, G., Tseng, D., Liao, C., Dorie, M. J., Czechowicz, A., Brown, J. M. 2010; 107 (18): 8363-8368

    Abstract

    Despite recent advances in radiotherapy, loco-regional failures are still the leading cause of death in many cancer patients. We have previously reported that bone marrow-derived CD11b(+) myeloid cells are recruited to tumors grown in irradiated tissues, thereby restoring the vasculature and tumor growth. In this study, we examined whether neutralizing CD11b monoclonal antibodies could inhibit the recruitment of myeloid cells into irradiated tumors and inhibit their regrowth. We observed a significant enhancement of antitumor response to radiation in squamous cell carcinoma xenografts in mice when CD11b antibodies are administered systemically. Histological examination of tumors revealed that CD11b antibodies reduced infiltration of myeloid cells expressing S100A8 and matrix metalloproteinase-9. CD11b antibodies further inhibited bone marrow-derived cell adhesion and transmigration to C166 endothelial cell monolayers and chemotactic stimuli, respectively, to levels comparable to those from CD11b knockout or CD18 hypomorphic mice. Given the clinical availability of humanized CD18 antibodies, we tested two murine tumor models in CD18 hypomorphic or CD11b knockout mice and found that tumors were more sensitive to irradiation when grown in CD18 hypomorphic mice but not in CD11b knockout mice. When CD18 hypomorphism was partially rescued by reconstitution with the wild-type bone marrow, the resistance of the tumors to irradiation was restored. Our study thus supports the rationale of using clinically available Mac-1 (CD11b/CD18) antibodies as an adjuvant therapy to radiotherapy.

    View details for DOI 10.1073/pnas.0911378107

    View details for Web of Science ID 000277310400058

    View details for PubMedID 20404138

    View details for PubMedCentralID PMC2889597

  • Purified Hematopoietic Stem Cell Transplantation: The Next Generation of Blood and Immune Replacement IMMUNOLOGY AND ALLERGY CLINICS OF NORTH AMERICA Czechowicz, A., Weissman, I. L. 2010; 30 (2): 159-?

    Abstract

    Replacement of disease-causing stem cells with healthy ones has been achieved clinically via hematopoietic cell transplantation (HCT) for the last 40 years, as a treatment modality for a variety of cancers and immunodeficiencies with moderate, but increasing, success. This procedure has traditionally included transplantation of mixed hematopoietic populations that include hematopoietic stem cells (HSC) and other cells, such as T cells. This article explores and delineates the potential expansion of this technique to treat a variety of inherited diseases of immune function, the current barriers in HCT and pure HSC transplantation, and the up-and-coming strategies to combat these obstacles.

    View details for DOI 10.1016/j.iac.2010.03.003

    View details for Web of Science ID 000279115200003

    View details for PubMedID 20493393

    View details for PubMedCentralID PMC3071240

  • Niche recycling through division-independent egress of hematopoietic stem cells JOURNAL OF EXPERIMENTAL MEDICINE Bhattacharya, D., Czechowicz, A., Ooi, A. G., Rossi, D. J., Bryder, D., Weissman, I. L. 2009; 206 (12): 2837-2850

    Abstract

    Hematopoietic stem cells (HSCs) are thought to reside in discrete niches through stable adhesion, yet previous studies have suggested that host HSCs can be replaced by transplanted donor HSCs, even in the absence of cytoreductive conditioning. To explain this apparent paradox, we calculated, through cell surface phenotyping and transplantation of unfractionated blood, that approximately 1-5% of the total pool of HSCs enters into the circulation each day. Bromodeoxyuridine (BrdU) feeding experiments demonstrated that HSCs in the peripheral blood incorporate BrdU at the same rate as do HSCs in the bone marrow, suggesting that egress from the bone marrow to the blood can occur without cell division and can leave behind vacant HSC niches. Consistent with this, repetitive daily transplantations of small numbers of HSCs administered as new niches became available over the course of 7 d led to significantly higher levels of engraftment than did large, single-bolus transplantations of the same total number of HSCs. These data provide insight as to how HSC replacement can occur despite the residence of endogenous HSCs in niches, and suggest therapeutic interventions that capitalize upon physiological HSC egress.

    View details for DOI 10.1084/jem.20090778

    View details for Web of Science ID 000272079300020

    View details for PubMedID 19887396

    View details for PubMedCentralID PMC2806613

  • Niche Recycling through Division-Independent Egress of Hematopoietic Stem Cells. 51st Annual Meeting and Exposition of the American-Society-of-Hematology Czechowicz, A., Bhattacharya, D., Ooi, L., Rossi, D. J., Bryder, D., Weissman, I. L. AMER SOC HEMATOLOGY. 2009: 37–37
  • Hematopoietic stem cell quiescence attenuates DNA damage response and permits DNA damage accumulation during aging CELL CYCLE Rossi, D. J., Seita, J., Czechowicz, A., Bhattacharya, D., Bryder, D., Weissman, I. L. 2007; 6 (19): 2371-2376

    Abstract

    The aging of tissue-specific stem and progenitor cells is believed to be central to the pathophysiological conditions arising in aged individuals. While the mechanisms driving stem cell aging are poorly understood, mounting evidence points to age-dependent DNA damage accrual as an important contributing factor. While it has been postulated that DNA damage may deplete stem cell numbers with age, recent studies indicate that murine hematopoietic stem cell (HSC) reserves are in fact maintained despite the accrual of genomic damage with age. Evidence suggests this to be a result of the quiescent (G0) cell cycle status of HSC, which results in an attenuation of checkpoint control and DNA damage responses for repair or apoptosis. When aged stem cells that have acquired damage are called into cycle under conditions of stress or tissue regeneration however, their functional capacity was shown to be severely impaired. These data suggest that age-dependent DNA damage accumulation may underlie the diminished capacity of aged stem cells to mediate a return to homeostasis after acute stress or injury. Moreover, the cytoprotection afforded by stem cell quiescence in stress-free, steady-state conditions suggests a mechanism through which potentially dangerous lesions can accumulate in the stem cell pool with age.

    View details for Web of Science ID 000251085700012

    View details for PubMedID 17700071

  • Adult human hematopoietic cells differentiate into mature T cells via a CD3-4+8-intermediate within the mouse thymic microenvironment; A new model system for the study of human thymocyte development. 47th Annual Meeting of the American-Society-of-Hematology Kraft, D. L., Czechowicz, A., Weissman, I. L. AMER SOC HEMATOLOGY. 2005: 155A–156A