Bio

Bio


Prof. Agnieszka Czechowicz is our newest and youngest faculty member within the Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine. Although a recent recruit, she previously spent a decade on the Farm as a Stanford undergraduate, medical student and graduate student and completed her PhD work with Prof. Irv Weissman one of the great leaders in stem cell biology. As a physician-scientist, Dr. Czechowicz subsequently did clinical training in Boston, completing her residency in Pediatrics at the prestigious Boston Children’s Hospital and pursued subspecialty training in Pediatric Hematology/Oncology at the Dana Farber Cancer Institute while simultaneously conducting postdoctoral research with Prof. Derrick Rossi and Prof. David Scadden. Her primary clinical interest is in bone marrow failure and aplastic anemia, and in other diseases commonly neccessitating stem cell transplantation.

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, and multiple pre-clinical and clinical therapies are in development based upon her studies. 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 Beth Ann Brown (bethannb@stanford.edu).

Clinical Focus


  • Pediatric Hematology-Oncology
  • Hematopoietic Stem Cell Transplantation
  • Bone Marrow Transplantation
  • Bone Marrow Failure
  • Inherited Genetic Diseases
  • Fanconi Anemia
  • Aplastic Anemia
  • Bone Marrow Metastasis
  • Bone Marrow Harvesting
  • Organ Tolerance
  • Pediatrics
  • Clinical Trials
  • Gene Therapy / Gene-Editing

Administrative Appointments


  • Member, Stanford Diabetes Research Center (2018 - Present)

Professional Education


  • Fellowship:Stanford University Pediatric Hematology Oncology Fellowship (2017) CA
  • Clinical Fellowship III, Stanford University, Pediatric Hematology/Oncology/Transplant (2017)
  • Clinical Fellowship I-II, Dana Farber Cancer Institute, Pediatric Hematology/Oncology/Transplant (2016)
  • Fellowship:Boston Children's Hospital Training Verifications (2017) MA
  • Clinical Residency, Boston Children's Hospital, Pediatrics (2014)
  • Residency:Boston Children's Hospital Training Verifications (2014) MA
  • Medical Education:Stanford University School of Medicine Registrar (2012) CA

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

Research & Scholarship

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. Currently Stanford has one open clinical trial derived from Dr. Czechowicz’s research for patients with severe combined immunodeficiency, and she is in the process of opening up additional clinical studies. https://clinicaltrials.gov/ct2/show/NCT02963064

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

Teaching

2018-19 Courses


Stanford Advisees


Publications

All Publications


  • 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

  • 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
  • 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

    View details for DOI 10.1111/petr.12309

    View details for Web of Science ID 000340530800017

  • 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 Web of Science ID 000315841900062

    View details for PubMedID 23382202

    View details for PubMedCentralID PMC3587280

  • 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

  • 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

  • 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