1 / 69

Cell, Tissue, and Gene Therapies

Cell, Tissue, and Gene Therapies. Elizabeth Read, MD May 11, 2011. Cell, Tissue & Gene Therapies. Heterogeneous group of (potential) products Very few products on the market Regulatory framework has evolved relatively recently (over past 20 years) Special development considerations.

bebe
Télécharger la présentation

Cell, Tissue, and Gene Therapies

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Cell, Tissue, andGene Therapies Elizabeth Read, MD May 11, 2011

  2. Cell, Tissue & Gene Therapies • Heterogeneous group of (potential) products • Very few products on the market • Regulatory framework has evolved relatively recently (over past 20 years) • Special development considerations

  3. Cell-based therapies originated with hematopoietic transplantation in 1970s • Bone marrow harvested, filtered, and transferred to blood bags in operating room • BM product carried directly to patient unit for infusion • Minimal donor & product testing, graft manipulation, quality systems • To date, FDA considers conventional autologous and allogeneic family-related BMT as “Practice of Medicine”

  4. 1980s – 2000s • Advances in science & technology spurred novel approaches for development of cell-based therapies • Hematopoietic transplants with “engineered” grafts starting with bone marrow, peripheral blood, or cord blood sources • Immunotherapies • T cells & subpopulations • Dendritic cell tumor vaccines • NK cells • Cellular gene therapies • Cell therapies derived from bone marrow, other tissues, and organs (e.g. mesenchymal stem cells, pancreatic islets)

  5. During this period, clinical translation was facilitated by development of technologies for collecting & handling cells in closed systems (often with single-use disposables)…

  6. And also by development of automated, large scale systems for cell collection, separation & isolation

  7. 2000s – PresentStem Cells & Regenerative Medicine • Explosion in stem cell science led to interest in use of stem cell-based therapies for many diseases and conditions, from cosmetic to life-threatening • Multipotent • Adult stem cells from bone marrow, fat & other tissues/organs • Fetal stem cells & placental stem cells are usually considered “adult” • Pluripotent • Embryonic stem (ES) cells • Induced pluripotent stem (iPS) cells

  8. Scope of cell & tissue therapies • Bone marrow and other hematopoietic stem cell transplantation • Cellular immunotherapies (dendritic cell vaccines, NK cells, T cells, etc) • Cell therapies derived from stem cells • Adult (including fetal) stem cells • Induced pluripotent stem cells • Embryonic stem cells • Cellular gene therapies • Conventional organ transplantation (e.g., kidney, heart, liver) • Conventional tissue transplantation (e.g., tendons, bone) • Reproductive tissue (sperm, oocytes, embryos) • Tissue engineering (autologous, allogeneic) – may include synthetic or natural biomaterials, or decellularized tissues • Xenotransplantation

  9. How does FDA regulate these products?

  10. Development pathway for cell & tissue therapies is similar to drugs & conventional biologics But with many important exceptions

  11. Exception #1 • Transplantation of vascularized whole organs is regulated by HRSA, not FDA

  12. Exception #2 • Xenotransplantation is regulated by its own separate set of FDA regulations

  13. Exception #3 • Bone marrow transplantation using autologous or family-related allogeneic donors is not regulated at all (practice of medicine)

  14. Exception #4 • Bone marrow transplantation from unrelated donors is regulated by HRSA, not FDA

  15. Exception #5 • Some tissue products have been regulated by CDRH as devices, with less stringent requirements and minimal involvement of CBER • This is historical – CBER will be involved going forward

  16. What’s left in scope? • Bone marrow and other hematopoietic stem cell transplantation • Cellular immunotherapies (dendritic cell vaccines, NK cells, T cells, etc) • Cell therapies derived from stem cells • Adult (including fetal) stem cells • Induced pluripotent stem cells • Embryonic stem cells • Cellular gene therapies • Conventional organ transplantation (e.g., kidney, heart, liver) • Conventional tissue transplantation (e.g., tendons, bone) • Reproductive tissue (sperm, oocytes, embryos) • Tissue engineering (autologous, allogeneic) – may include synthetic or natural biomaterials, or decellularized tissues • Xenotransplantation

  17. What’s left falls into FDA definition ofHCT/Ps • Human cells, tissues, and cellular and tissue-based products (HCT/Ps) are articles containing human cells or tissues that are intended for implantation, transplantation, infusion, or transfer into a human recipient

  18. FDA’s Risk-Based Approach forHCT/Ps • Lower risk “361” • Autologous or family related donors and minimally manipulated and homologous use • Regulated under section 361 of Public Health Service Act • Higher risk “351” • Allogeneic unrelated donors and/or more than minimally manipulated and/or non-homologous use • Regulated under section 351 of Public Health Service Act, and subject to same rules as drugs & other biologics for IND and premarket approval

  19. FDA regulations for HCT/Ps

  20. What about stem cells? Cellular products derived from multipotent or pluripotent stem cells are regulated as HCT/Ps

  21. HCT/Ps derived from pluripotent stem cells: FDA concerns • CMC • Donor source • Consistency of differentiation & expansion process • Detection of residual pluripotent stem cells • Genetic and epigenetic stability • Preclinical studies • Case-by-case approach • “hybrid” efficacy/safety studies – much attention to modeling ROA and biodistribution • Tumorigenicity

  22. HCT/Ps derived from pluripotent stem cells: FDA concerns • Clinical Protocol: for novel stem cell products, the risk : benefit assessment is difficult; therefore: • Rationale for clinical trial must be justified by especially strong proof of concept • Greater emphasis placed on product characterization and preclinical testing

  23. Gene Therapies

  24. Gene therapy approaches • IN VIVO: Vector administered directly to patient, and transfers genetic information to patient cells in vivo • Intravenously administered vector delivers gene for factor IX to patient with hemophilia B • EX VIVO: Vector used to transfer genetic information to cells ex vivo, then cells are administered to patient • Vector that delivers gene for enzyme adenosine deaminase is incubated ex vivo with autologous lymphocytes of patient with ADA-deficient form of SCID (severe combined immunodeficiency), and genetically modified cells are infused to patient

  25. Gene therapy: history • 1974: NIH established Recombinant DNA Advisory Committee (RAC) • NIH Guidelines on recombinant DNA research • 1980s: New subcommittee of RAC to oversee clinical gene therapy • Appendix M to NIH Guidelines – covered design of preclinical & clinical research, consent issues, AE reporting • PUBLIC review of gene transfer protocols • 1989: First clinical gene transfer study (gene marking) using retroviral vector • 1990: First clinical gene transfer study (therapeutic intent) using retroviral vector

  26. Gene therapy: history • 1995: No real clinical efficacy demonstrated, and NIH report concluded that enthusiasm had outstripped knowledge • Back to the bench for research on improved gene delivery methods (e.g., higher titer vectors, use of stromal feeder layer or fibronectin for HSC transductions) • By 1995, NIH RAC • Had approved 149 GT clinical protocols • No dire consequences • Policy change: public review & approval only for GT protocols that presented novel or unresolved issues • 1997: Role of NIH RAC modified – still required public review, but not “approval” of novel GT protocols

  27. Jessie Gelsinger (1999) • 18 y.o. with clinically mild form of ornithine transcarbamlase defiency • Volunteered for clinical trial of gene therapy at U of Pennsylvania • Adenoviral vector caused massive immune response, muti-organ failure, and death within 4 days • All gene therapy trials placed on hold • Multiple ethical issues raised • Adverse events in primate studies • Adverse events in 2 previous human subjects • Informed consent • Principal investigator conflict of interest

  28. Insertional Oncogenesis • 2000-2007: X-linked SCID trials, using gamma retroviral vectors to deliver the corrective gene (IL2RG) to autologous hematopoietic progenitor cells • 5 of 20 pts developed T cell leukemia-like proliferative disorder, caused by INSERTIONAL ONCOGENESIS • Retroviral vector integrated adjacent to one or more cellular proto-oncogenes (LMO-2 in 4 of the cases), which increased their expression, leading to malignant transformation and outgrowth of clonal population of T cells

  29. Gene delivery methods • Vector = an agent used to introduce genetic material into cells • Vectors can be • Viral • Non-viral • Plasmid DNA • Liposomes or other agents that facilitate entry into cell

  30. Viral vectors • Retrovirus and lentivirus (developed to overcome inability of γ-retroviral vectors to infect non-dividing cells) • Adenovirus • Parvovirus (adeno-associated virus or AAV) • Herpes simplex virus • Poxvirus • Togavirus

  31. Vector selection depends on… • Disease state • Route of administration • Size of payload • genetic sequences, regulatory elements • Cell cycling • Lentivirus, adenovirus, AAV do not require cycling cells • Intended duration of expression • Retrovirus and lentivirus give stable integration • Plasmid used for transient expression • Target cells • Poor expression of adenoviral CAR receptor on hematopoietic cells

  32. More advanced vector design features • Conditional replication-competence • Control of gene expression • Tissue-specific promoters • Drug-responsive promoters • To reduce risk of insertional oncogenesis ofγ-retroviral and lentiviral vectors • Self-inactivating (SIN design) • Insulators • Suicide genes • Ganciclovir administered to patient will kill cells with thymidine kinase gene

  33. Safety issues • Observed to date • Insertional mutagenesis/oncogenesis • Immunogenicity • Vector • Transgene • FBS (bovine protein used to manufacture vector) • Potential • Inadvertent transmission & expression in non-target cells (including germline, transplacental)

  34. FDA regulations & guidance for gene therapies • Overall similar to biotechnology products • ICH guidances • Gene therapy CMC guidance 2008 • Vector description, map, sequence analysis • Cell banks, viral banks, cell lines (packaging, producer, feeder) • Vector production/purification • Documentation of RAC review • For ex vivo gene therapy, cell requirements same as HCT/Ps (i.e. CMC guidance, tissue rules)

  35. FDA guidance on GT delayed AEs • Recommends preclinical study designs to assess clinical risk • Requires long term clinical follow up, based on preclinical studies, for • In vivo gene therapy with persistence of vector sequences, when sequences are integrated • Ex vivo gene therapy with sequences integrated, or not integrated but have potential for latency & reactivation • Specific follow up observations yearly for at least 10 years, and reporting to FDA • Informed consent for long term follow up, and for use of retroviral vectors

  36. RCR/RCL testing(FDA 2006 supplemental guidance)

  37. Case Study Cellular gene therapy for sickle cell disease PI - Donald Kohn MD (UCLA) Funded by CIRM

  38. Sickle Cell Disease (SCD) • Autosomal recessive disorder • Approx 8% of African Americans have mutation • Approx 1 in 500 African Americans is homozygous and has SCD • Clinical course • hemolytic anemia • vaso-occlusive episodes (pain), strokes, acute chest syndrome, progressive organ dysfunction

  39. Molecular basis of SCD • Substitution of T for A in 6th codon of human β-globin gene • Results in non-polar valine instead of polar glutamic acid on the surface of HbS tetramer (α2βS2)

  40. Molecular basis of SCD • During partial deoxygenation, valine creates hydrophobic pocket that fits into natural hydrophilic pocket on HbS tetramers, leading to HbS polymerization • This causes red blood cells to become rigid and poorly deformable, leading to hemolysis and impaired blood flow through microcirculation

  41. Treatment of SCD • Supportive for vaso-occlusive crisis • Pain medication, hydration, oxygen • Blood transfusions • For some acute complications • Prophylaxis for stroke and other complications • Complications: iron overload, alloimmunization • Hydroxyurea • Key mechanism: raises Hb F, which has anti-sickling effect • Complications: pancytopenia • Allogenic bone marrow transplantation • Potential for cure, but only 14% have HLA-matched sibling donor

  42. Potential Gene Therapy Strategies for SCD • Correct HbS mutation • But sickle β-globin acts in a dominant manner, and you would need very high levels of expression to achieve a state similar to sickle trait • Insert genes for normal HbF γ-globin into HSCs, in order to increase expression of HbF (α2γ2), to inhibit Hb S polymerization and sickling • But fetal γ-globin gene is poorly expressed in adult RBCs, due to absence of fetal-specific positive regulatory factors in adult cells • Modify HbS β-globin gene to have anti-sickling properties of γ-globin while retaining the adult HSC expression pattern inherent in the β-globin gene

  43. Townes βAS3 vector • Self-inactivating (SIN) lentiviral vector • Carries and expressesβAS3, a β-globin gene with 3 amino acid substitutions • Expression product has biophysical anti-sickling properties equivalent to fetal γ-globin AND advantage over βS–globin for dimerization with α-globin • Incorporates β-globin transcriptional regulatory elements

  44. Preclinical Proof of Concept(Levasseur 2003) • In murine model of SCD, transduction of HSC with the lenti/βAS3 vector • Expression: 2-3 gm Hb/dl/vector copy • Correction of hematological and clinical manifestations of SCD

  45. IND development for SCD gene therapy

  46. Clinical protocol considerations • Phase 1 trial • Risks: known and unknown • Benefits: unlikely in first trial • SCD patient population • Adults (ethical considerations for children) • Should not be candidates for allo BMT (i.e., matched sibling donor available) • Severity of disease may impact • feasibility of cell collection • endpoint assessment • Myeloablation with busulfan to create “space” in marrow

  47. Product considerations • Vector: based on Townes SIN lentiviral vector • Additional engineering underway to further reduce risk of insertional oncogenesis • TAT independent backbone, insulators, etc. • Cell source • Autologous • Ideally want most primitive hematopoietic stem cells (HSCs) that will differentiate into erthyroid cells • HSCs vs iPS cells • iPS cells not quite ready for prime time • HSCs have track record, CD34+ selection isolates stem & progenitor cells

  48. Product considerations • HSC options • Placental/umbilical cord blood • most proliferative source, but not useful for autologous protocol in adults • G-CSF mobilized peripheral blood HSCs • SCD patients have had serious adverse events, including death, associated with G-CSF • Bone marrow • Will require general anesthesia • Available cell dose will be an issue

  49. Initial definition of product candidate • The investigational product is autologous human CD34+ hematopoietic stem cells (HSC) from the bone marrow of patients with sickle cell disease (SCD) modified by ex vivo transduction using the βAS3 lentiviral vector

More Related