Preclinical development and qualification of ZFN-mediated CCR5 disruption in human hematopoietic stem/progenitor cells

David L DiGiusto, Paula M Cannon, Michael C Holmes, Lijing Li, Anitha Rao, Jianbin Wang, Gary Lee, Philip D Gregory, Kenneth A Kim, Samuel B Hayward, Kathleen Meyer, Colin Exline, Evan Lopez, Jill Henley, Nancy Gonzalez, Victoria Bedell, Rodica Stan, John A Zaia, David L DiGiusto, Paula M Cannon, Michael C Holmes, Lijing Li, Anitha Rao, Jianbin Wang, Gary Lee, Philip D Gregory, Kenneth A Kim, Samuel B Hayward, Kathleen Meyer, Colin Exline, Evan Lopez, Jill Henley, Nancy Gonzalez, Victoria Bedell, Rodica Stan, John A Zaia

Abstract

Gene therapy for HIV-1 infection is a promising alternative to lifelong combination antiviral drug treatment. Chemokine receptor 5 (CCR5) is the coreceptor required for R5-tropic HIV-1 infection of human cells. Deletion of CCR5 renders cells resistant to R5-tropic HIV-1 infection, and the potential for cure has been shown through allogeneic stem cell transplantation with naturally occurring homozygous deletion of CCR5 in donor hematopoietic stem/progenitor cells (HSPC). The requirement for HLA-matched HSPC bearing homozygous CCR5 deletions prohibits widespread application of this approach. Thus, a strategy to disrupt CCR5 genomic sequences in HSPC using zinc finger nucleases was developed. Following discussions with regulatory agencies, we conducted IND-enabling preclinical in vitro and in vivo testing to demonstrate the feasibility and (preclinical) safety of zinc finger nucleases-based CCR5 disruption in HSPC. We report here the clinical-scale manufacturing process necessary to deliver CCR5-specific zinc finger nucleases mRNA to HSPC using electroporation and the preclinical safety data. Our results demonstrate effective biallelic CCR5 disruption in up to 72.9% of modified colony forming units from adult mobilized HSPC with maintenance of hematopoietic potential in vitro and in vivo. Tumorigenicity studies demonstrated initial product safety; further safety and feasibility studies are ongoing in subjects infected with HIV-1 (NCT02500849@clinicaltrials.gov).

Figures

Figure 1
Figure 1
Evaluation of the effects of electroporation of HSPC with rSB-728mR. (a) Extent of CCR5 disruption as estimated by (open box) MiSeq and (closed box) Surveyor nuclease assay (Cel-1) after electroporation with varying concentrations of research grade CCR5-specific ZFN mRNA (rSB-728mR). Regarding to MiSeq data, ~ 3,000–30,000 total sequence reads per sample were obtained and used for calculation of % CCR5 disruption (% indels). (b) Extent of modification of CCR5 and next four top off-target sequences after electroporation with varying concentrations of rSB-728mR. (c) Viability of HSPC on day 1 (D1) and day 2 (D2) after electroporation (EP). (d) The effects of electroporation on hematopoietic potential measured as colony forming units (CFU) of HSPC plated 2 days after electroporation with 50 or 150 μg/ml rSB-728mR or150 μg/ml GMP Grade SB-728 mRNA. Controls were treated with or without 150 μg/ml rSB-728mR but without electroporation (No EP). A total of 500 HSPC were plated from each sample, and CFUs were read at 14 days. *P < 0.05, **P < 0.01. CCR5, chemokine receptor 5; HSPCs, hematopoietic stem/progenitor cells; ZFNs, zinc finger nucleases.
Figure 2
Figure 2
Engraftment studies. CD34+ cells from two healthy donors were stimulated overnight and subjected to electroporation using 150 µg/ml of the GMP grade mRNA (SB-728mR). (a) The %CD45+ cells in bone marrow and spleen, analyzed at 20 weeks, are shown with error bars. The frequency (%) of the engrafted human CD45+ cells that were CD14+ monocytes, CD19+ B cells, CD4+ T cells and CD8+ T cells in bone marrow (b) and in spleen (c) are shown with error bars. *P < 0.05.
Figure 3
Figure 3
On-target and off-target sequence disruption. CD34+ cells were electroporated with 150 µg/ml SB-728mR and placed in bulk culture or methyl cellulose for CFU assay, as described in “Materials and Methods”. (a) Bulk cultures and single colonies were analyzed by miSeq and the percent of bulk or total CFU modified, percent of CFU with biallelic modifications among all CFU and percent of CFU with biallelic among all modified CFU is shown (b) The frequency of disruption among on/off target sequences are shown. CFU, colony forming units.
Figure 4
Figure 4
SB-728mR has no impact on HSPC engraftment. Cohorts of NSG mice were transplanted with 106 HSPC per mouse from three independent donors (GLP#1, GLP#3, and GLP#4) following electroporation in the presence of 150 µg/ml SB-728mR or no electroporation and followed for percent CD45+ cells in peripheral blood at (a) 4-, 12, and 22-weeks and (b) in bone marrow (BM) at necropsy. *P < 0.05; **P < 0.01. Dotted line shows minimal threshold for defining engraftment with human cells. Note: BM samples were lost from one control and one SB-728mR-HSPC animal due to operator error. All week-22 GLP#4 control blood samples and 8 of 25 SB-728mR-HSPC blood samples were lost due to technical issues. HSPCs, hematopoietic stem/progenitor cells.
Figure 5
Figure 5
Percent CCR5 gene modification in mice transplanted with SB-728mR HSPC. NSG mice were transplanted with 106 HSPC from three independent donors (GLP#1, GLP#3, and GLP#4) following electroporation (EP) in the presence of 150 µg/ml SB-728mR or no electroporation (No EP) and followed for percent CD45+ cells in peripheral blood mononuclear cells at 4-, 12-, and 22-weeks. CCR5, chemokine receptor 5; HSPCs, hematopoietic stem/progenitor cells.

References

    1. DiGiusto, DL, Stan, R, Krishnan, A, Li, H, Rossi, JJ and Zaia, JA (2013). Development of hematopoietic stem cell based gene therapy for HIV-1 infection: considerations for proof of concept studies and translation to standard medical practice. Viruses 5: 2898–2919.
    1. Tebas, P, Stein, D, Tang, WW, Frank, I, Wang, SQ, Lee, G et al. (2014). Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med 370: 901–910.
    1. Raport, CJ, Gosling, J, Schweickart, VL, Gray, PW and Charo, IF (1996). Molecular cloning and functional characterization of a novel human CC chemokine receptor (CCR5) for RANTES, MIP-1beta, and MIP-1alpha. J Biol Chem 271: 17161–17166.
    1. Deng, H, Liu, R, Ellmeier, W, Choe, S, Unutmaz, D, Burkhart, M et al. (1996). Identification of a major co-receptor for primary isolates of HIV-1. Nature 381: 661–666.
    1. Dragic, T, Litwin, V, Allaway, GP, Martin, SR, Huang, Y, Nagashima, KA et al. (1996). HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 381: 667–673.
    1. Doranz, BJ, Rucker, J, Yi, Y, Smyth, RJ, Samson, M, Peiper, SC et al. (1996). A dual-tropic primary HIV-1 isolate that uses fusin and the beta-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell 85: 1149–1158.
    1. Choe, H, Farzan, M, Sun, Y, Sullivan, N, Rollins, B, Ponath, PD et al. (1996). The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85: 1135–1148.
    1. Feng, Y, Broder, CC, Kennedy, PE and Berger, EA (1996). HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 272: 872–877.
    1. Liu, R, Paxton, WA, Choe, S, Ceradini, D, Martin, SR, Horuk, R et al. (1996). Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 86: 367–377.
    1. Zimmerman, PA, Buckler-White, A, Alkhatib, G, Spalding, T, Kubofcik, J, Combadiere, C et al. (1997). Inherited resistance to HIV-1 conferred by an inactivating mutation in CC chemokine receptor 5: studies in populations with contrasting clinical phenotypes, defined racial background, and quantified risk. Mol Med 3: 23–36.
    1. Allers, K, Hütter, G, Hofmann, J, Loddenkemper, C, Rieger, K, Thiel, E et al. (2011). Evidence for the cure of HIV infection by CCR5Δ32/Δ32 stem cell transplantation. Blood 117: 2791–2799.
    1. Yukl, SA, Boritz, E, Busch, M, Bentsen, C, Chun, TW, Douek, D et al. (2013). Challenges in detecting HIV persistence during potentially curative interventions: a study of the Berlin patient. PLoS Pathog 9: e1003347.
    1. Krishnan, A and Zaia, JA (2014). HIV-associated non-Hodgkin lymphoma: viral origins and therapeutic options. Hematology Am Soc Hematol Educ Program 2014: 584–589.
    1. Holt, N, Wang, J, Kim, K, Friedman, G, Wang, X, Taupin, V et al. (2010). Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol 28: 839–847.
    1. Li, L, Krymskaya, L, Wang, J, Henley, J, Rao, A, Cao, LF et al. (2013). Genomic editing of the HIV-1 coreceptor CCR5 in adult hematopoietic stem and progenitor cells using zinc finger nucleases. Mol Ther 21: 1259–1269.
    1. Perez, EE, Wang, J, Miller, JC, Jouvenot, Y, Kim, KA, Liu, O et al. (2008). Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 26: 808–816.
    1. Chabot, S, Rosazza, C, Golzio, M, Zumbusch, A, Teissié, J and Rols, MP (2013). Nucleic acids electro-transfer: from bench to bedside. Curr Drug Metab 14: 300–308.
    1. Van Tendeloo, VF, Ponsaerts, P and Berneman, ZN (2007). mRNA-based gene transfer as a tool for gene and cell therapy. Curr Opin Mol Ther 9: 423–431.
    1. Morgan, RA and Kakarla, S (2014). Genetic modification of T cells. Cancer J 20: 145–150.
    1. Li, L, Allen, C, Shivakumar, R and Peshwa, MV (2013). Large volume flow electroporation of mRNA: clinical scale process. Methods Mol Biol 969: 127–138.
    1. Watanabe, M, Umeyama, K, Matsunari, H, Takayanagi, S, Haruyama, E, Nakano, K et al. (2010). Knockout of exogenous EGFP gene in porcine somatic cells using zinc-finger nucleases. Biochem Biophys Res Commun 402: 14–18.
    1. Doyon, Y, Choi, VM, Xia, DF, Vo, TD, Gregory, PD and Holmes, MC (2010). Transient cold shock enhances zinc-finger nuclease-mediated gene disruption. Nat Methods 7: 459–460.
    1. Gabriel, R, Lombardo, A, Arens, A, Miller, JC, Genovese, P, Kaeppel, C et al. (2011). An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat Biotechnol 29: 816–823.
    1. DiGiusto, DL, Krishnan, A, Li, L, Li, H, Li, S, Rao, A et al. (2010). RNA-based gene therapy for HIV with lentiviral vector-modified CD34(+) cells in patients undergoing transplantation for AIDS-related lymphoma. Sci Transl Med 2: 36ra43.
    1. Campbell, JH, Hearps, AC, Martin, GE, Williams, KC and Crowe, SM (2014). The importance of monocytes and macrophages in HIV pathogenesis, treatment, and cure. AIDS 28: 2175–2187.
    1. Kumar, A, Abbas, W and Herbein, G (2014). HIV-1 latency in monocytes/macrophages. Viruses 6: 1837–1860.
    1. Honeycutt, JB, Wahl, A, Baker, C, Spagnuolo, RA, Foster, J, Zakharova, O et al. (2016). Macrophages sustain HIV replication in vivo independently of T cells. J Clin Invest 126: 1353–1366.
    1. McNamara, LA and Collins, KL (2011). Hematopoietic stem/precursor cells as HIV reservoirs. Curr Opin HIV AIDS 6: 43–48.
    1. McNamara, LA, Ganesh, JA and Collins, KL (2012). Latent HIV-1 infection occurs in multiple subsets of hematopoietic progenitor cells and is reversed by NF-κB activation. J Virol 86: 9337–9350.
    1. Keating, GM (2011). Plerixafor: a review of its use in stem-cell mobilization in patients with lymphoma or multiple myeloma. Drugs 71: 1623–1647.
    1. Yannaki, E, Anagnostopoulos, A, Karponi, G, Zervou, F, Constantinou, V, Psatha, N et al. (2013). Superior Stem Cell Mobilization by a Combination of Plerixafor + G-CSF in Patients with Thalassemia. In: American Society of Gene & Cell Therapy 16th Annual Meeting. American Society for Cell & Gene Therapy Salt Lake City, Utah.
    1. Vose, JM, Ho, AD, Coiffier, B, Corradini, P, Khouri, I, Sureda, A et al. (2009). Advances in mobilization for the optimization of autologous stem cell transplantation. Leuk Lymphoma 50: 1412–1421.
    1. Alexander, ET, Towery, JA, Miller, AN, Kramer, C, Hogan, KR, Squires, JE et al. (2011). Beyond CD34+ cell dose: impact of method of peripheral blood hematopoietic stem cell mobilization (granulocyte-colony-stimulating factor [G-CSF], G-CSF plus plerixafor, or cyclophosphamide G-CSF/granulocyte-macrophage [GM]-CSF) on number of colony-forming unit-GM, engraftment, and Day +100 hematopoietic graft function. Transfusion 51: 1995–2000.
    1. Donahue, RE, Jin, P, Bonifacino, AC, Metzger, ME, Ren, J, Wang, E et al. (2009). Plerixafor (AMD3100) and granulocyte colony-stimulating factor (G-CSF) mobilize different CD34+ cell populations based on global gene and microRNA expression signatures. Blood 114: 2530–2541.
    1. Brave, M, Farrell, A, Ching Lin, S, Ocheltree, T, Pope Miksinski, S, Lee, SL et al. (2010). FDA review summary: Mozobil in combination with granulocyte colony-stimulating factor to mobilize hematopoietic stem cells to the peripheral blood for collection and subsequent autologous transplantation. Oncology 78: 282–288.
    1. Miller, DT, Adam, MP, Aradhya, S, Biesecker, LG, Brothman, AR, Carter, NP et al. (2010). Consensus statement: chromosomal microarray is a first-tier clinical diagnostic test for individuals with developmental disabilities or congenital anomalies. Am J Hum Genet 86: 749–764.
    1. Boring, L, Gosling, J, Chensue, SW, Kunkel, SL, Farese, RV Jr, Broxmeyer, HE et al. (1997). Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J Clin Invest 100: 2552–2561.
    1. Lim, JK, Obara, CJ, Rivollier, A, Pletnev, AG, Kelsall, BL and Murphy, PM (2011). Chemokine receptor Ccr2 is critical for monocyte accumulation and survival in West Nile virus encephalitis. J Immunol 186: 471–478.
    1. Lin, KL, Sweeney, S, Kang, BD, Ramsburg, E and Gunn, MD (2011). CCR2-antagonist prophylaxis reduces pulmonary immune pathology and markedly improves survival during influenza infection. J Immunol 186: 508–515.
    1. Lim, JK, Louie, CY, Glaser, C, Jean, C, Johnson, B, Johnson, H et al. (2008). Genetic deficiency of chemokine receptor CCR5 is a strong risk factor for symptomatic West Nile virus infection: a meta-analysis of 4 cohorts in the US epidemic. J Infect Dis 197: 262–265.

Source: PubMed

3
Abonneren