Provided herein are genetically engineered hematopoietic cells such as hematopoietic stem cells having one or more genetically edited genes of cell-surface proteins, that can be used in combination with immunotherapies (i.e., cytotoxic agents, such as chimeric antigen receptor T cells), and therapeutic uses thereof.
Innovations in gene transfer have made it possible to reprogram immune cells to target molecules expressed on cancer cells. Exceptionally promising results led the FDA to approve the first adoptive cellular immunotherapy, known as CD19 CAR-T cells, for the treatment of B lymphoblastic leukemia. Although these successes are expected to revolutionize the oncology field, their application has been hampered because most suitable candidates are often shared with healthy bone marrow cells, leading to immunosuppression and severe hematopoietic toxicity. Anti-myeloid/stem cell CAR-T-induced toxicity restricts their applicability to a salvage therapy in a limited time window before HSCT, which may be insufficient for disease eradication. Thus, there remains an unmet need to effectively target cells of interest, e.g., cancer cells, without targeting or harming normal cell populations.
The present disclosure generally relates to genetically engineered hematopoietic cells such as hematopoietic stem cells having one or more genetically edited genes of cell-surface proteins, and chimeric antigen receptors that are capable of targeting the same cell-surface proteins.
In one embodiment, provided is a genetically engineered hematopoietic stem cell (HSPC), comprising a genetically engineered FLT3 gene, wherein the genetically engineered FLT3 gene is engineered such that its encoded protein has reduced binding to a therapeutic anti-FLT3 antibody. In an embodiment, the genetically engineered FLT3 gene comprises at least one mutation in exon 9 of the FLT3 gene. In an embodiment, at least one mutation in exon 9 of the genetically engineered FLT3 gene results in a polypeptide bearing a mutation at position N399. In an embodiment, the mutation at position N399 is N399D or N399G. In an embodiment, the therapeutic anti-FLT3 antibody is anti-FLT3 clone 4G8 antibody. In an embodiment, the therapeutic anti-FLT3 antibody is an antibody that has the same six CDRs as, or competes with, 4G8 antibody. In an embodiment, the genetically engineered HSPCs are genetically engineered using a CRISPR system comprising a guide nucleic acid and a nuclease. In an embodiment, the nuclease is either Streptococcus pyogenes Cas9 (SpCas9), Staphylococcus aureus (SaCas9), Lachnospiraceae bacterium Cas12a (LbCas12a), or Acidaminococcus sp. BV3L6 (AsCas12a). In one embodiment, the CRISPR system comprises SpCas9. In an embodiment, the guide nucleic acid is selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16. In an embodiment, CRISPR system further comprises a template DNA. In an embodiment, the template DNA is selected from the group consisting of SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, and SEQ ID NO: 43. In an embodiment, the nuclease is a catalytically impaired SpCas9 linked to a base editor enzyme. In an embodiment, the base editor enzyme is a nucleotide deaminase. In an embodiment, the nucleotide deaminase is either a cytosine deaminase or an adenosine deaminase. In another embodiment, the catalytically impaired SpCas9 is NG-SpCas9 or SpRY-SpCas9. In an embodiment, the catalytically impaired SpCas9 comprises a mutation at position D10A. In an embodiment, the catalytically impaired SpCas9 further comprises a mutation at position K918N. In an embodiment, the guide RNA is selected from the group consisting of SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 23. In an embodiment, the genetically engineered FLT3 gene encodes a polypeptide which comprises the amino acid sequence of SEQ ID NO: 51 or SEQ ID NO: 52. Also provided is a population of genetically engineered hematopoietic stem cells (HSPCs), comprising the genetically engineered HSPCs as described above. In some embodiments, provided is a method of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) the population of genetically engineered hematopoietic stem cells as described above, and (b) a therapeutically effective amount of at least one agent comprising an anti-FLT3 antibody binding domain or an antibody or antibody fragment comprising the anti-FLT3 antibody binding domain. In an embodiment, the at least one agent comprises a Chimeric Antigen Receptor-T (CAR-T) cell comprising the anti-FLT3 antibody binding domain. In an embodiment, the hematopoietic malignancy is B-lymphoblastic leukemia (BLL), acute myeloid leukemia (AML), or T-cell acute lymphoblastic leukemia (T-ALL). In an embodiment, the method further comprises obtaining HSPCs from a biological sample from the human subject and genetically engineering the HSPCs from the biological sample from the human subject, thereby forming the population of genetically engineered HSPCs. In an embodiment, the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
Also provided in an embodiment, is a genetically engineered hematopoietic stem cell (HSPC), comprising a genetically engineered CD123 gene, wherein the genetically engineered CD123 gene is engineered such that its encoded protein has reduced binding to a therapeutic anti-CD123 antibody. In an embodiment, the therapeutic anti-CD123 antibody is clone 7G3 antibody or its humanized counterpart CSL362. In an embodiment, the genetically engineered CD123 gene comprises at least one mutation in exon 2 of the CD123 gene. In an embodiment, at least one mutation in exon 2 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position S59. In an embodiment, the mutation at S59 is S59P or S59F. In an embodiment, the therapeutic anti-CD123 antibody is anti-CD123 clone 6H6 antibody or anti-CD123 clone S18016F antibody. In an embodiment, the genetically engineered CD123 gene comprises at least one mutation in exon 3 of the CD123 gene. In an embodiment, at least one mutation in exon 3 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position P88. In an embodiment, the mutation at P88 is P88L or P88S. In an embodiment, the genetically engineered HSPCs are genetically engineered using a CRISPR system comprising a guide nucleic acid and a nuclease. In an embodiment, the guide nucleic acid is selected from the group consisting of SEQ ID NO: 24, SEQ ID NO: 27, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34. In an embodiment, the nuclease is a catalytically impaired SpCas9 linked to a base editor enzyme. In an embodiment, the base editor enzyme is a nucleotide deaminase. In an embodiment, the base editor enzyme is either a cytosine deaminase or an adenosine deaminase. In an embodiment, the catalytically impaired SpCas9 is NG-SpCas9 or SpRY-SpCas9. In an embodiment, the catalytically impaired SpCas9 comprises a mutation at position D10A. In an embodiment, the catalytically impaired SpCas9 further comprises a mutation at position K918N. In an embodiment, the genetically engineered CD123 gene encodes a polypeptide which comprises the amino acid sequence of SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, or SEQ ID NO: 58. Also provided herein is a population of genetically engineered hematopoietic stem cells (HSPCs), comprising the genetically engineered HSPCs as described above. Also provided is a method of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) the population of genetically engineered hematopoietic stem cells as described above, and (b) a therapeutically effective amount of at least one agent comprising an anti-CD123 antibody binding domain or an antibody or antibody fragment comprising the anti-CD123 binding domain. In an embodiment, the at least one agent comprises a Chimeric Antigen Receptor-T (CAR-T) cell comprising the anti-CD123 antibody binding domain. In an embodiment, the hematopoietic malignancy is B-lymphoblastic leukemia (BLL), acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or Blastic Plasmacytoid Dendritic Cell Leukemia (BPCDN). In an embodiment, the method further comprises obtaining HSPCs from a biological sample from the human subject and genetically engineering the HSPCs from the biological sample from the human subject, thereby forming the population of genetically engineered HSPCs. In an embodiment, the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
In some embodiments provided is a population of genetically engineered hematopoietic stem cells (HSPCs) comprising: (i) a genetically engineered FLT3 gene, wherein the genetically engineered FLT3 gene encodes a protein that has reduced binding to a therapeutic anti-FLT3 antibody, and (ii) a genetically engineered CD123 gene, wherein the genetically engineered CD123 gene encodes a protein that has reduced binding to a therapeutic anti-CD123 antibody. In an embodiment, the genetically engineered FLT3 gene comprises at least one mutation in exon 9 of the FLT3 gene. In an embodiment, at least one mutation in exon 9 of the genetically engineered FLT3 gene results in a polypeptide bearing a mutation at position N399. In an embodiment, the genetically engineered CD123 gene comprises at least one mutation in exon 2 of the CD123 gene. In an embodiment, at least one mutation in exon 2 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position S59. In an embodiment, the therapeutic anti-FLT3 antibody is anti-FLT3 clone 4G8 antibody. In an embodiment, the therapeutic anti-CD123 antibody is anti-CD123 clone 7G3 antibody or CSL362 antibody. In an embodiment, the population of HSPCs are genetically engineered using a CRISPR system comprising at least two guide nucleic acids and a nuclease. In an embodiment, the at least two guide nucleic acids are 1) SEQ ID NO: 18 or SEQ ID NO: 20 and 2) SEQ ID NO: SEQ ID NO: 24 or SEQ ID NO: 27. In an embodiment, the at least two guide nucleic acids are SEQ ID NO: 20 and SEQ ID NO: 27. In an embodiment, the nuclease is a catalytically impaired SpCas9 linked to a base editor enzyme. In an embodiment, the base editor enzyme is a nucleotide deaminase. In an embodiment, the base editor enzyme is either a cytosine deaminase or an adenosine deaminase. In an embodiment, the catalytically impaired SpCas9 is NG-SpCas9 or SpRY-SpCas9. In an embodiment, the catalytically impaired SpCas9 comprises a mutation at position D10A. In an embodiment, the SpCas9 further comprises a mutation at position K918N. Also provided is a method of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) the population of HSPCs as described above, and (b) a therapeutically effective amount of at least one agent comprising one or both of: (1) an anti-FLT3 antibody binding domain or an antibody or antibody fragment comprising the anti-FLT3 antibody binding domain, and/or (2) an anti-CD123 antibody binding domain or an antibody or antibody fragment comprising the anti-CD123 binding domain. In an embodiment, the at least one agent comprises a Chimeric Antigen Receptor-T (CAR-T) cell comprising the anti-FLT3 antibody binding domain and/or the anti-CD123 antibody binding domain. In an embodiment, the hematopoietic malignancy is B-lymphoblastic leukemia (BLL), acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or Blastic Plasmacytoid Dendritic Cell Leukemia (BPCDN). In an embodiment, the method further comprises obtaining HSPCs from a biological sample from the human subject and genetically engineering the HSPCs from the biological sample from the human subject, thereby forming the population of genetically engineered HSPCs. In an embodiment, the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
Also provided in an embodiment is a genetically engineered hematopoietic stem cell (HSPC), comprising a genetically engineered KIT gene, wherein the genetically engineered KIT gene is engineered such that its encoded protein has reduced binding to a therapeutic anti-KIT antibody. In an embodiment, the genetically engineered KIT gene comprises at least one mutation in exon 7 of the KIT gene. In an embodiment, at least one mutation in exon 7 of the genetically engineered KIT gene results in a polypeptide bearing a mutation at position H378. In an embodiment, the mutation at position H378 is H378R. In an embodiment, the therapeutic anti-KIT antibody is anti-KIT clone Fab79D antibody. In an embodiment, the genetically engineered HSPCs are genetically engineered using a CRISPR system comprising a guide nucleic acid and a nuclease. In an embodiment, the guide nucleic acid is selected from the group consisting of SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, and SEQ ID NO: 39. In an embodiment, the nuclease is a catalytically impaired SpCas9 linked to a base editor enzyme. In an embodiment, wherein the base editor enzyme is a nucleotide deaminase. In an embodiment, the base editor enzyme is either a cytosine deaminase or an adenosine deaminase. In an embodiment, the catalytically impaired SpCas9 is NG-SpCas9 or SpRY-SpCas9. In an embodiment, the catalytically impaired SpCas9 comprises a mutation at position D10A. In an embodiment, SpCas9 further comprises a mutation at position K918N. Also provided is a population of genetically engineered hematopoietic stem cells (HSPCs), comprising the genetically engineered HSPCs as described above. Also provided is a method of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) the population of genetically engineered hematopoietic stem cells as described above, and (b) a therapeutically effective amount of at least one agent comprising the anti-KIT antibody binding domain or an antibody or antibody fragment comprising the anti-KIT binding domain. In an embodiment, the at least one agent comprises a Chimeric Antigen Receptor-T (CAR-T) cell comprising the anti-KIT antibody binding domain. In an embodiment, the hematopoietic malignancy is B-lymphoblastic leukemia (BLL), acute myeloid leukemia (AML), or T-cell acute lymphoblastic leukemia (T-ALL). In an embodiment, the method further comprising obtaining HSPCs from a biological sample from the human subject and genetically engineering the HSPCs from the biological sample from the human subject, thereby forming the population of genetically engineered HSPCs. In an embodiment, the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
Also provided in some embodiments is a population of genetically engineered hematopoietic stem cells (HSPCs) comprising: (i) a genetically engineered KIT gene, wherein the genetically engineered KIT gene encodes a protein that has reduced binding to a therapeutic anti-KIT antibody, and (ii) a genetically engineered CD123 gene, wherein the genetically engineered CD123 gene encodes a protein that has reduced binding to a therapeutic anti-CD123 antibody. In an embodiment, the genetically engineered KIT gene comprises at least one mutation in exon 7 of the KIT gene. In an embodiment, at least one mutation in exon 7 of the genetically engineered KIT gene results in a polypeptide bearing a mutation at position H378. In an embodiment, the genetically engineered CD123 gene comprises at least one mutation in exon 2 of the CD123 gene. In an embodiment, at least one mutation in exon 2 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position S59. In an embodiment, the therapeutic anti-KIT antibody is anti-KIT clone Fab79D antibody. In an embodiment, the therapeutic anti-CD123 antibody is anti-CD123 clone 7G3 antibody or CSL362 antibody. In an embodiment, the population of HSPCs are genetically engineered using a CRISPR system comprising at least two guide nucleic acids and a nuclease. In an embodiment, the at least two guide nucleic acids are 1) SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, or SEQ ID NO: 39 and 2) SEQ ID NO: 24 or SEQ ID NO: 27. In an embodiment, the at least two guide nucleic acids are SEQ ID NO: 37 and SEQ ID NO: 27. In an embodiment, the nuclease is a catalytically impaired SpCas9 linked to a base editor enzyme. In an embodiment, the base editor enzyme is a nucleotide deaminase. In an embodiment, the base editor enzyme is either a cytosine deaminase or an adenosine deaminase. In an embodiment, the catalytically impaired SpCas9 is NG-SpCas9 or SpRY-SpCas9. In an embodiment, the catalytically impaired SpCas9 comprises a mutation at position D10A. In an embodiment, the SpCas9 further comprises a mutation at position K918N. Also provided is a method of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) the population of HSPCs as described above, and (b) a therapeutically effective amount of at least one agent comprising one or both of: (1) an anti-KIT antibody binding domain or an antibody or antibody fragment comprising the anti-KIT binding domain, and/or (2) an anti-CD123 antibody binding domain or an antibody or antibody fragment comprising the anti-CD123 binding domain. In an embodiment, the at least one agent comprises a Chimeric Antigen Receptor-T (CAR-T) cell comprising the anti-KIT antibody binding domain and/or the anti-CD123 antibody binding domain. In an embodiment, the hematopoietic malignancy is B-lymphoblastic leukemia (BLL), acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or Blastic Plasmacytoid Dendritic Cell Leukemia (BPCDN). In an embodiment, the method further comprises obtaining HSPCs from a biological sample from the human subject and genetically engineering the HSPCs from the biological sample from the human subject, thereby forming the population of genetically engineered HSPCs. In an embodiment, the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
Also provided herein is a population of genetically engineered hematopoietic stem cells (HSPCs) comprising: (i) a genetically engineered FLT3 gene, wherein the genetically engineered FLT3 gene encodes a protein that has reduced binding to a therapeutic anti-FLT3 antibody, and (ii) a genetically engineered KIT gene, wherein the genetically engineered KIT gene encodes a protein that has reduced binding to a therapeutic anti-KIT antibody. In an embodiment, the genetically engineered FLT3 gene comprises at least one mutation in exon 9 of the FLT3 gene. In an embodiment, at least one mutation in exon 9 of the genetically engineered FLT3 gene results in a polypeptide bearing a mutation at position N399. In an embodiment, the genetically engineered KIT gene comprises at least one mutation in exon 7 of the KIT gene. In an embodiment, at least one mutation in exon 7 of the genetically engineered KIT gene results in a polypeptide bearing a mutation at position H378. In an embodiment, the therapeutic anti-FLT3 antibody is anti-FLT3 clone 4G8 antibody. In an embodiment, the therapeutic anti-KIT antibody is anti-KIT clone Fab79D antibody. In an embodiment, the population of HSPCs are genetically engineered using a CRISPR system comprising at least two guide nucleic acids and a nuclease. In an embodiment, the at least two guide nucleic acids are 1) SEQ ID NO: 18 or SEQ ID NO: 20 and 2) SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, or SEQ ID NO: 39. In an embodiment, the at least two guide nucleic acids are SEQ ID NO: 20 and SEQ ID NO: 37. In an embodiment, the nuclease is a catalytically impaired SpCas9 linked to a base editor enzyme. In an embodiment, the base editor enzyme is a nucleotide deaminase. In an embodiment, the base editor enzyme is either a cytosine deaminase or an adenosine deaminase. In an embodiment, the catalytically impaired SpCas9 is NG-SpCas9 or SpRY-SpCas9. In an embodiment, the catalytically impaired SpCas9 comprises a mutation at position D10A. In an embodiment, the SpCas9 further comprises a mutation at position K918N. Also provided is a method of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) the population of HSPCs as described above, and (b) a therapeutically effective amount of at least one agent comprising one or both of: (1) an anti-FLT3 antibody binding domain or an antibody or antibody fragment comprising the anti-FLT3 antibody binding domain, and/or (2) an anti-KIT antibody binding domain or an antibody or antibody fragment comprising the anti-KIT binding domain. In an embodiment, the at least one agent comprises a Chimeric Antigen Receptor-T (CAR-T) cell comprising the anti-FLT3 antibody binding domain and/or the anti-KIT antibody binding domain. In an embodiment, the hematopoietic malignancy is B-lymphoblastic leukemia (BLL), acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL, or Blastic Plasmacytoid Dendritic Cell Leukemia (BPCDN). In an embodiment, the method further comprises obtaining HSPCs from a biological sample from the human subject and genetically engineering the HSPCs from the biological sample from the human subject, thereby forming the population of genetically engineered HSPCs. In an embodiment, the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
Also provided in some embodiments is a chimeric antigen receptor (CAR) comprising a polypeptide comprising: (a) one or more epitope binding fragments that binds to an epitope of one or more cell-surface lineage-specific proteins, (b) a hinge domain, (c) a transmembrane domain, (d) a co-stimulatory domain, and (e) a cytoplasmic signaling domain, wherein the one or more cell-surface lineage-specific proteins are selected from FLT3, CD123, and/or KIT.
In some embodiments, the cell-surface lineage-specific protein is FLT3 and the CAR comprises the amino acid sequence of SEQ ID NO: 73. In some embodiments, the cell-surface lineage-specific protein is FLT3 and the one or more epitope binding fragments comprises the one or more epitope binding fragments from SEQ ID NO: 73. In some embodiments, the cell-surface lineage-specific protein is FLT3 and the one or more epitope binding fragments comprise the following CDR sequences: GYTFTSYWMH (SEQ ID NO: 96), EIDPSDSYKDYNQKFK (SEQ ID NO: 97), RAITTTPFDF (SEQ ID NO: 98), RASQSISNNLH (SEQ ID NO: 99), YASQSIS (SEQ ID NO: 100), and QQSNTWPYT (SEQ ID NO: 101). In some embodiments, the cell-surface lineage-specific protein is CD123 and the CAR comprises the amino acid sequence of SEQ ID NO: 75, SEQ ID NO: 86, or SEQ ID NO: 87. In some embodiments, the cell-surface lineage-specific protein is CD123 and the one or more epitope binding fragments comprises the one or more epitope binding fragments from SEQ ID NO: 75, SEQ ID NO: 86, or SEQ ID NO: 87. In some embodiments, the cell-surface lineage-specific protein is CD123 and the one or more epitope binding fragments comprise the following CDR sequences: GYSFTDYYMK (SEQ ID NO: 104), DIIPSNGATFYNQKFKG (SEQ ID NO: 105), ARSHLLRASWFAY (SEQ ID NO: 106), SQSLLNSGNQKNYLT (SEQ ID NO: 107), WASTRES (SEQ ID NO: 108), and QNDYSYPYT (SEQ ID NO: 109). In some embodiments, the cell-surface lineage-specific protein is CD123 and the one or more epitope binding fragments comprise the following CDR sequences: DIIPSNGATFYNQKFKG (SEQ ID NO: 105), SQSLLNSGNQKNYLT (SEQ ID NO: 107), WASTRES (SEQ ID NO: 108), and QNDYSYPYT (SEQ ID NO: 109). In some embodiments, the cell-surface lineage-specific protein is KIT and the CAR comprises the amino acid sequence of SEQ ID NO: 69 or SEQ ID NO: 71. In some embodiments, the cell-surface lineage-specific protein is KIT and the one or more epitope binding fragments comprises the one or more epitope binding fragments from SEQ ID NO: 69 or SEQ ID NO: 71. In some embodiments, the cell-surface lineage-specific protein is KIT and the one or more epitope binding fragments comprise the following CDR sequences: GFNISVYMMH (SEQ ID NO: 88), SIYPYSGYTYYADSVKG (SEQ ID NO: 89), ARYVYHALDY (SEQ ID NO: 90), RASQRGLRNVAVA (SEQ ID NO: 91), SASSLYS (SEQ ID NO: 92), and QQWAVHSLIT (SEQ ID NO: 93). In some embodiments, the one or more cell-surface lineage-specific proteins are FLT3 and CD123 and the CAR comprises the amino acid sequence of SEQ ID NO: 77 or SEQ ID NO: 79. In some embodiments, the one or more cell-surface lineage-specific proteins are FLT3 and CD123 and the one or more epitope binding fragments comprises the one or more epitope binding fragments from SEQ ID NO: 77 or SEQ ID NO: 79. In some embodiments, the one or more cell-surface lineage-specific proteins are FLT3 and CD123 and the one or more epitope binding fragments comprise the following CDR sequences: GYTFTSYWMH (SEQ ID NO: 96), EIDPSDSYKDYNQKFK (SEQ ID NO: 97), RAITTTPFDF (SEQ ID NO: 98), RASQSISNNLH (SEQ ID NO: 99), YASQSIS (SEQ ID NO: 100), QQSNTWPYT (SEQ ID NO: 101), GYSFTDYYMK (SEQ ID NO: 104), DIIPSNGATFYNQKFKG (SEQ ID NO: 105), ARSHLLRASWFAY (SEQ ID NO: 106), SQSLLNSGNQKNYLT (SEQ ID NO: 107), WASTRES (SEQ ID NO: 108), and QNDYSYPYT (SEQ ID NO: 109). In some embodiments of any of the CARs described above, the hinge domain is a CD28 hinge, an IgG4 hinge, or a CD8α hinge. In some embodiments of any of the CARs described above, the transmembrane domain is a CD28 TM, a CD8α TM, or a 4-1BB™. In some embodiments of any of the CARs described above, the co-stimulatory domain is CD28z, 4-1BB, ICOS, or OX40. In some embodiments of any of the CARs described above, wherein the cytoplasmic signaling domain is CD3z.
Also provided herein are cells expressing any of the above-described CARs. In some embodiments, the cell is an immune cell. In some embodiments, the immune cell is a T-cell.
Also provided herein are methods of treating a hematopoietic malignancy, the method comprising administering to a human subject: (a) a population of genetically engineered hematopoietic stem cells (such as any of those described above, and (b) cells expressing any of the above-described CARs (such as an immune cell).
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 51, wherein the polypeptide sequence comprises a mutation at N399D and wherein the polypeptide sequence has reduced binding to a therapeutic anti-FLT3 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 52, wherein the polypeptide sequence comprises a mutation at N399G and wherein the polypeptide sequence has reduced binding to a therapeutic anti-FLT3 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 54, wherein the polypeptide sequence comprises a mutation at S59P and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 55, wherein the polypeptide sequence comprises mutations at Y58H and S59P and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 56, wherein the polypeptide sequence comprises a mutation at S59F and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 57, wherein the polypeptide sequence comprises a mutation at P88S and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 58, wherein the polypeptide sequence comprises a mutation at P88L and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 67, wherein the polypeptide sequence comprises mutations at F316S, M318V, I319K, V323I, I334V, E360K, P363V, E366D, E376Q, and H378R and wherein the polypeptide sequence has reduced binding to a therapeutic anti-KIT antibody.
Also provided herein is a polypeptide sequence comprising a polypeptide sequence that is at least 80% identical to the sequence set forth in SEQ ID NO: 68, wherein the polypeptide sequence comprises a mutation at H378R and wherein the polypeptide sequence has reduced binding to a therapeutic anti-KIT antibody.
Also provided herein are nucleic encoding any of the above described polypeptides.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
Identifying suitable proteins for targeted cancer therapies presents a significant challenge. Many potential target proteins are present on both the cell surface of a cancer cell and on the cell surface of normal, non-cancer cells, which can be required or critically involved in the development and/or survival of the subject. Many of the target proteins contribute to the functionality of such essential cells. Thus, therapies targeting these proteins can lead to deleterious effects in the subject, such as significant toxicity and/or other side effects. Further, resistance to CAR-T therapy remains a challenge in treatment of hematopoietic malignancies, such as acute myeloid leukemia (AML) due to switch of cancer antigens on cancer cells, thereby escaping CAR-T therapy.
Accordingly, the present disclosure provides methods, cells, compositions, and kits aimed at addressing at least the above-stated problems. The methods, cells, compositions, and kits described herein provide a safe and effective treatment for hematological malignancies, allowing for targeting of one or more cell surface proteins that are present not only on cancer cells but also on cells critical for the development and/or survival of the subject. In some instances, described herein are genetically engineered hematopoietic cells such as hematopoietic stem cells (HSPCs) having genetic editing in one or more genes coding for cell-surface proteins, for example, FLT3, CD123, and/or KIT; methods of producing such, for examples, via the CRISPR approach using specific guide RNAs; methods of treating a hematopoietic malignancy using the engineered hematopoietic cells, either taken alone, or in combination with one or more cytotoxic agents (e.g., CAR-T cells) that can target the wild-type cell-surface antigens but not those encoded by the edited genes in the engineered hematopoietic cells; and kits comprising the engineered hematopoietic cells.
In some embodiments, the genetically engineered hematopoietic cells have an edited FLT3 gene, CD123 gene, or KIT gene. In some in, one or more of these genes are mutated. In some instances, the mutated FLT3 gene, CD123 gene, or KIT gene include mutations or deletions in one or more non-essential epitopes so as to retain (in whole or in part) the bioactivity of FLT3 gene, CD123 gene, or KIT gene.
i. Hematopoietic Stem Cells
In some embodiments, the hematopoietic cells described herein are hematopoietic stem cells. Hematopoietic stem/progenitor cells (HSPCs) are capable of giving rise to both myeloid and lymphoid progenitor cells that further give rise to myeloid cells (e.g., monocytes, macrophages, neutrophils, basophils, dendritic cells, erythrocytes, platelets, etc.) and lymphoid cells (e.g., T cells, B cells, NK cells), respectively. HSPCs are characterized by the expression of the cell surface marker CD34 (e.g., CD34+), which can be used for the identification and/or isolation of HSPCs.
In some embodiments, the HSPCs are obtained from a subject, such as a mammalian subject. In some embodiments, the mammalian subject is a non-human primate, a rodent (e.g., mouse or rat), a bovine, a porcine, an equine, or a domestic animal. In some embodiments, the HSPCs are obtained from a human patient, such as a human patient suffering from a hematopoietic malignancy. In some embodiments, the HSPCs are obtained from a healthy donor. In some embodiments, the HSPCs are obtained from the subject to whom the genetically engineered HSPCs will be subsequently administered. HSPCs that are administered to the same subject from which the cells were obtained are referred to as autologous cells, whereas HSPCs that are obtained from a subject who is not the subject to whom the cells will be administered are referred to as allogeneic cells (methods to reduce incidence of rejection are standard and well known in the art).
HSPCs can be obtained from any suitable source using convention means known in the art. In some embodiments, HSPCs are obtained from a sample from a subject (or donor), such as bone marrow sample or from a blood sample. Alternatively or in addition, HSPCs can be obtained from an umbilical cord (i.e. cord blood cells). In some embodiments, the HSPCs are from bone marrow, cord blood cells, or peripheral blood mononuclear cells (PBMCs). In general, bone marrow cells can be obtained from iliac crest, femora, tibiae, spine, rib or other medullary spaces of a subject (or donor). Bone marrow can be taken out of the patient and isolated through various separations and washing procedures known in the art. An exemplary procedure for isolation of bone marrow cells comprises the following steps: a) extraction of a bone marrow sample; b) centrifugal separation of bone marrow suspension in three fractions and collecting the intermediate fraction, or buffy coat; c) the buffy coat fraction from step (b) is centrifuged one more time in a separation fluid, commonly Ficoll™, and an intermediate fraction which contains the bone marrow cells is collected; and d) washing of the collected fraction from step (c) for recovery of re-transfusable bone marrow cells.
HSPCs typically reside in the bone marrow but can be mobilized into the circulating blood by administering a mobilizing agent in order to harvest HSPCs from the peripheral blood. In some embodiments, the subject (or donor) from which the HSPCs are obtained is administered a mobilizing agent, such as granulocyte colony-stimulating factor (G-CSF). The number of the HSPCs collected following mobilization using a mobilizing agent is typically greater than the number of cells obtained without use of a mobilizing agent.
In some embodiments, a sample is obtained from a subject (or donor) and is then enriched for a desired cell type (e.g. CD34+, CD34+CD38−, CD133+, CD90+, CD49f+). For example, PBMCs and/or CD34+ hematopoietic cells can be isolated from blood as described herein. Cells can also be isolated from other cells, for example by isolation and/or activation with an antibody binding to an epitope on the cell surface of the desired cell type. Another method that can be used includes negative selection using antibodies to cell surface markers to selectively enrich for a specific cell type without activating the cell by receptor engagement.
ii. Mutated Cell-Surface Antigens
In some embodiments, the hematopoietic stem cells (HSPCs) described herein can contain an edited gene encoding one or more cell-surface proteins of interest (e.g. FLT3, CD123, KIT) in mutated form (mutants or variants, which are used herein interchangeably), which has reduced binding or no binding to a cytotoxic agent as described herein (e.g. anti-FLT3 antibody, anti-CD123 antibody, anti-KIT antibody). The mutants can carry one or more mutations of the epitope to which the cytotoxic agent binds, such that binding to the cytotoxic agent is reduced or abolished as compared to the natural or wild-type cell-surface protein counterpart. Such a mutant is preferred to maintain substantially similar biological activity as the wild-type counterpart.
As used herein, the term “reduced binding” refers to binding that is reduced by at least 25%. The level of binding can refer to the amount of binding of the cytotoxic agent to a hematopoietic stem cell or the amount of binding of the cytotoxic agent to the cell-surface protein as compared to a wild-type (i.e., non-engineered, non-mutated) protein. In some embodiments, the binding is reduced by at least 25%, 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. In some embodiments, the binding is reduced such that there is substantially no detectable binding in a conventional assay. As used herein, “no binding” refers to substantially no binding, e.g., no detectable binding or only baseline binding as determined in a conventional binding assay.
In some instances, the variant contains one or more amino acid residue substitutions (e.g., 2, 3, 4, 5, or more) within the epitope of interest such that the cytotoxic agent does not bind or has reduced binding to the mutated epitope. Such a mutant can have substantially reduced binding affinity to the cytotoxic agent (e.g., having a binding affinity that is at least 40%, 50%, 60%, 70%, 80% or 90% lower than its wild-type counterpart). In some examples, such a variant can have abolished binding activity to the cytotoxic agent. In other instances, the mutant contains a deletion of a region that comprises the epitope of interest. Such a region can be encoded by an exon. In some embodiments, the region is a domain of the cell-surface protein of interest that encodes the epitope. In one example, the variant has just the epitope deleted. The length of the deleted region can range from 3-60 amino acids, e.g., 5-50, 5-40, 10-30, 10-20, etc.
The mutation(s) or deletions in a mutant of a cell-surface antigen can be within or surround a non-essential epitope such that the mutation(s) or deletion(s) do not substantially affect the bioactivity of the protein.
As used herein, the term “epitope” refers to an amino acid sequence (linear or conformational) of a protein, such as a cell-surface antigen, that is bound by the CDRs of an antibody. In some embodiments, the cytotoxic agent binds to one or more (e.g., at least 2, 3, 4, 5 or more) epitopes of a cell-surface antigen. In some embodiments, the cytotoxic agent binds to more than one epitope of the cell-surface antigen and the hematopoietic cells are manipulated such that each of the epitopes is absent and/or unavailable for binding by the cytotoxic agent.
In some embodiments, the genetically engineered HSPCs described herein have one or more edited genes of cell-surface antigens such that the edited genes express mutated cell-surface antigens with mutations in one or more non-essential epitopes. A “non-essential epitope” (or a fragment comprising such) refers to a domain within the cell surface protein/antigen, the mutation in which is less likely to substantially affect the bioactivity of the cell surface protein. For example, hematopoietic cells comprising a deletion or mutation of a non-essential epitope of a cell-surface antigen, such hematopoietic cells are able to proliferate and/or undergo erythropoietic differentiation to a similar level as hematopoietic cells that express a wild-type cell-surface antigen.
Methods for identifying and/or verifying non-essential epitopes in cell-surface antigens would be known and recognized by one of ordinary skill in the art and is also within the scope of the present disclosure. Further, methods for assessing the functionality of the cell-surface antigen and the hematopoietic cells are known in the art and include, for example, proliferation assays, differentiation assays, colony formation, expression analysis (e.g., gene and/or protein), protein localization, intracellular signaling, functional assays, and in vivo humanized mouse models.
iii. Preparation of Genetically Engineered Hematopoietic Cells
Any of the genetically engineering hematopoietic cells, such as HSPCs, that carry edited genes of one or more cell-surface antigens can be prepared by a routine method or by a method described herein. In some embodiments, the genetic engineering is performed using genome editing. As used herein, “genome editing” refers to a method of modifying the genome, including any protein-coding or non-coding nucleotide sequence, of an organism to alter the expression of a target gene. In general, genome editing methods involve use of an endonuclease that is capable of cleaving the nucleic acid of the genome, for example at a targeted nucleotide sequence. In some instances, genome editing methods involve use of a dead nuclease or a nuclease that is a nickase. Repair of the double-stranded breaks in the genome can be repaired introducing mutations and/or exogenous nucleic acid can be inserted into the targeted site. In some cases, genome editing methods involve use of a catalytically inactive or partially inactive endonuclease fused to a functional domain, e.g. an adenine or cytidine deaminase domain in the case of base editors. Other functional domains include prime editors, CRISPR-Cas activators or repressors, etc.
Genome editing methods are generally classified based on the type of endonuclease that is involved in generating double stranded breaks in the target nucleic acid. These methods include use of zinc finger nucleases (ZFN), transcription activator-like effector-based nuclease (TALEN), meganucleases, and CRISPR/Cas systems.
In one aspect of the present disclosure, the replacement of cancer cells by a modified population of normal cells is performed using normal cells that have been manipulated such that the cells do not bind the cytotoxic agent. Such modification can include the deletion or mutation of an epitope of the specific cell-surface protein using a CRISPR-Cas system, where the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas system is an engineered, non-naturally occurring CRISPR-Cas system.
CRISPR systems encode RNA-guided endonucleases that are essential for bacterial adaptive immunity. CRISPR-associated (Cas) nucleases can be readily programmed to cleave target DNA sequences for genome editing in various. One class of these nucleases, referred to as Cas9 proteins, complex with two short RNAs: a crRNA and a trans-activating crRNA (tracrRNA). The most commonly used Cas9 ortholog, SpCas9, uses a crRNA that has 20 nucleotides (nt) at its 5′ end that are complementary to the “protospacer” region of the target DNA site. Efficient cleavage also requires that SpCas9 recognizes a protospacer adjacent motif (PAM). The crRNA and tracrRNA are usually combined into a single ˜100-nt guide RNA (gRNA) that directs the DNA cleavage activity of SpCas9. A Cas protein named Cpf1 has been identified that can also be programmed to cleave target DNA sequences. Unlike SpCas9, Cpf1 requires only a single 42-nt crRNA, which has 23 nt at its 3′ end that are complementary to the protospacer of the target DNA sequence.
In some embodiments, the Cas endonuclease is a Cas9 nuclease or variant thereof. Cas9 endonucleases cleave double stranded DNA of a target nucleic acid resulting in blunt ends. In some embodiments, the Cas endonuclease is a Cpf1 nuclease or variant thereof. Cleavage with Cpf1 nucleases results in staggered ends of the nucleic acid.
In some embodiments, the Cas endonuclease is a Cas9 enzyme or variant thereof. In some embodiments, the Cas9 endonuclease is derived from Streptococcus pyogenes (SpCas9) or Staphylococcus aureus (SaCas9).
The SpCas9 wild type sequence is as follows (SEQ ID NO: 1):
In general, the target nucleic acid is flanked on the 3′ side or 5′ side by a protospacer adjacent motif (PAM) that can interact with the endonuclease and be further involved in targeting the endonuclease activity to the target nucleic acid. It is generally thought that the PAM sequence flanking the target nucleic acid depends on the endonuclease and the source from which the endonuclease is derived. For example, for Cas9 endonucleases that are derived from Streptococcus pyogenes, the PAM sequence is NGG, although the PAM sequences NAG and NGA can be recognized with lower efficiency. For Cas9 endonucleases derived from Staphylococcus aureus, the PAM sequence is NNGRRT (SEQ ID NO: 3).
Accordingly, in some instances, the endonuclease is engineered/modified such that it can recognize one or more PAM sequence. In some embodiments, the endonuclease has been engineered/modified to recognize one or more PAM sequence that is different than the PAM sequence the endonuclease recognizes without engineering/modification. In some embodiments, the endonuclease has been engineered/modified to reduce off-target activity of the enzyme. In some embodiments, the nucleotide sequence encoding the endonuclease is modified to alter the PAM recognition of the endonuclease. For example, the Cas endonuclease (e.g., SpCas9) has mutations at one or more of the following positions: A61, L1111, D1135, S1136, G1218, E1219, N1317, A1322, R1333, R1335, T1337. See, for example, International Patent Application Publication Nos. WO 2016/141224 and WO 2017/040348, US Patent Application Publication No. 2021/0284978A1, all of which are all incorporated herein by reference.
In some embodiments, the Cas9 endonuclease is the wild-type version of the nuclease. For example, the Cas9 endonuclease is an SpCas9 endonuclease having the sequence shown above in SEQ ID NO: 1. In some embodiments, the SpCas9 endonuclease is at least 80%, e.g., at least 85%, 90%, or 95% identical to the amino acid sequence of SEQ ID NO: 1, e.g., have differences at up to 5%, 10%, 15%, or 20% of the residues of SEQ ID NO: 1 replaced, e.g., with conservative mutations. In preferred embodiments, the endonuclease retains desired activity of the parent, e.g., the nuclease activity (except where the parent is a nickase or a dead Cas9), and/or the ability to interact with a guide RNA and target DNA). In other instances, the Cas9 endonuclease is an SaCas9 endonuclease having the sequence shown above in SEQ ID NO: 2. In some embodiments, the SaCas9 endonuclease is at least 80%, e.g., at least 85%, 90%, or 95% identical to the amino acid sequence of SEQ ID NO: 2, e.g., have differences at up to 5%, 10%, 15%, or 20% of the residues of SEQ ID NO: 2 replaced, e.g., with conservative mutations. In preferred embodiments, the endonuclease retains desired activity of the parent, e.g., the nuclease activity (except where the parent is a nickase or a dead Cas9), and/or the ability to interact with a guide RNA and target DNA).
In some embodiments, the Cas9 endonuclease is a catalytically inactive Cas9. For example, dCas9 contains mutations at catalytically active residues (D10, E762, D839, H983, or D986; and/or at H840 or N863) and does not have nuclease activity. For example, the mutations are: (i) D10A or D10N, and/or (ii) H840A, H840N, or H840Y.
In some embodiments, the Cas9 endonuclease includes a mutation at K918. For instance the mutation is K918N.
In some embodiments, the nucleotide sequence encoding the Cas9 endonuclease is further modified to alter the activity of the protein. In some embodiments, the Cas9 endonuclease has been modified to inactivate one or more catalytic residues of the endonuclease. In some embodiments, the Cas9 endonuclease has been modified to inactivate one of the catalytic residues of the endonuclease, referred to as a “nickase” or “Cas9n.” Cas9 nickase endonucleases cleave one DNA strand of the target nucleic acid.
In some instances, the endonuclease is a NG-SpCas9 nickase and has the following mutations: D10A, L1111R, D1135V, G1218R, E1219F, A1322R, R1335V, T1337R (relative to wild-type SpCas9). In some instances, the endonuclease is SpRY-Cas9 nickase and has the following mutations: D10A, A61R, L1111R, D1135L, S1136W, G1218K, E1219Q, N1317R, A1322R, R1333P, R1335Q, T1337R (relative to wild-type SpCas9).
In some embodiments, the Cas endonuclease is a Cpf1 nuclease or variant thereof. Cpf1 endonuclease generally recognizes a PAM sequence located at the 5′ end of the target nucleic acid. For a Cpf1 nuclease, the PAM sequence is TTTN. As will be appreciated by one of skill in the art, the Cas endonuclease Cpf1 nuclease can also be referred to as Cas12a. In some embodiments, the host cell expresses a Cpf1 nuclease derived from Lachnospiraceae bacterium (LbCpf1), Acidaminococcus sp. (AsCpf1), or Francisella tularensis (FnCpf1). Wild-type sequences for each are shown below.
In some embodiments, the Cpf1 endonuclease is the wild-type version of the nuclease. For example, the Cpf1 endonuclease is a Cpf1 endonuclease having the sequence shown above in SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6. In some embodiments, the Cpf1 endonuclease is at least 80%, e.g., at least 85%, 90%, or 95% identical to the amino acid sequence of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6, e.g., have differences at up to 5%, 10%, 15%, or 20% of the residues of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6 replaced, e.g., with conservative mutations. In preferred embodiments, the endonuclease retains desired activity of the parent, e.g., the nuclease activity (except where the parent is a nickase or a dead Cas9), and/or the ability to interact with a guide RNA and target DNA).
A catalytically inactive variant of Cpf1 (Cas12a) can be referred to dCas12a. Thus, in some embodiments, for AsCpf1, catalytic activity-destroying mutations are made at D908 and E993, e.g., D908A and E993A; and for LbCpf1 catalytic activity-destroying mutations at D832 and E925, e.g., D832A and E925A; and for FnCpf1 catalytic activity-destroying mutations at D917A and E1006A.
Alternatively or in addition, the Cas endonuclease (i.e., Cas9 or Cas12a) can be fused to another protein or portion thereof, e.g., a heterologous functional domain. In some embodiments, the heterologous functional domain is a transcriptional activation domain (e.g., VP64 or NF-KB p65). In some embodiments, the heterologous functional domain is a transcriptional silencer or transcriptional repression domain (e.g., wherein the transcriptional repression domain is Krueppel-associated box (KRAB) domain, ERF repressor domain (ERD), or mSin3A interaction domain (SID); wherein the transcriptional silencer is Heterochromatin Protein 1 (HP1)). In some embodiments, the heterologous functional domain is an enzyme that modifies the methylation state of DNA (e.g., a DNA methyltransferase (DNMT) or a TET protein (such as, TET1)). In some embodiments, the heterologous functional domain is an enzyme that modifies a histone subunit (e.g., a histone acetyltransferase (HAT), histone deacetylase (HDAC), histone methyltransferase (HMT), or histone demethylase). In some embodiments, the heterologous functional domain is a biological tether (e.g., MS2, Csy4 or lambda N). In some embodiments, the heterologous functional domain is FokI.
In some embodiments, the heterologous functional domain is a base editor, such as a deaminase that modifies cytosine DNA bases, e.g., a cytidine deaminase from the apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like (APOBEC) family of deaminases, including APOBEC1, APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D/E, APOBEC3F, APOBEC3G, APOBEC3H, APOBEC4, activation-induced cytidine deaminase (AID), cytosine deaminase 1 (CDA1), and CDA2, and cytosine deaminase acting on tRNA (CDAT). Specific examples include, evoAPOBEC1-BE4max, eA3A-BE5, and EA-BE4max.
In some embodiments, the heterologous functional domain is a deaminase that modifies adenosine DNA bases, e.g., the deaminase is an adenosine deaminase 1 (ADA1), ADA2; adenosine deaminase acting on RNA 1 (ADAR1), ADAR2, ADAR3; adenosine deaminase acting on tRNA 1 (ADAT1), ADAT2, ADAT3; and naturally occurring or engineered tRNA-specific adenosine deaminase (TadA). For example, ABE8e-TadA-8e. In some embodiments, the TadA adenosine deaminase domain includes a V106W substitution.
In some embodiments, the heterologous functional domain is an enzyme, domain, or peptide that inhibits or enhances endogenous DNA repair or base excision repair (BER) pathways, e.g., uracil DNA glycosylase inhibitor (UGI) that inhibits uracil DNA glycosylase (UDG, also known as uracil N-glycosylase, or UNG) mediated excision of uracil to initiate BER; or DNA end-binding proteins such as Gam from the bacteriophage Mu.
In some embodiments, the endonuclease is a base editor. Base editor endonuclease generally comprises a catalytically inactive Cas endonuclease fused to a base editor. For example, the endonuclease is SpCas9 with a mutation at D10, E762, D839, H983, or D986; and/or at H840 or N863 and fused to a base editor, such as those mentioned above.
In some instances, the endonuclease (Cas9 or Cas12a) is fused to one or more of a nuclear localization sequence, cell penetrating peptide sequence, affinity tag, and/or a fluorescent protein. For example, the nuclear localization sequence is the SV40 large T-antigen nuclear localization sequence (PKKKRKV; SEQ ID NO: 82), the nucleoplasmin nuclear localization sequence (KRPAATKKAGQAKKKK; SEQ ID NO: 83) or the c-Myc nuclear localization sequence (PAAKRVKLD; SEQ ID NO: 84). For example, the nuclear localization sequence(s) is fused to the N-terminus and/or to the C-terminus of the Cas9 or Cas12a protein. In some embodiments, when a heterologous functional domain is fused to the N-terminus and/or to the C-terminus of the Cas9 or Cas12a protein, the nuclear localization sequence(s) is fused to the N-terminus and/or to the C-terminus of the heterologous functional domain-Cas protein complex or interposed between the heterologous functional domain and the Cas protein.
Sequences of exemplary Cas endonucleases are provided below:
gRNA
The terms “gRNA,” “guide RNA” and “CRISPR guide sequence” can be used interchangeably throughout and refer to a nucleic acid comprising a sequence that determines the specificity of a Cas DNA binding protein of a CRISPR/Cas system. A gRNA hybridizes to (complementary to, partially or completely) a target nucleic acid sequence in the genome of a host cell. In some instances, the gRNA refers collectively to the crRNA and the tracrRNA (for instance, when a Cas9 nuclease is being used—in those instances, the guide RNA may be referred to as a single guide RNA, i.e., sgRNA). In other instances, the gRNA refers only to the crRNA (for instance, when a Cpf1 endonuclease is being used). The gRNA or portion thereof that hybridizes to the target nucleic acid can be between 15-25 nucleotides, 18-22 nucleotides, or 19-21 nucleotides in length. In some embodiments, the gRNA sequence that hybridizes to the target nucleic acid is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length. In some embodiments, the gRNA sequence that hybridizes to the target nucleic acid is between 10-30, or between 15-25, nucleotides in length. In some embodiments, the gRNA sequence is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or at least 100% complementary to a target nucleic acid.
Exemplary guide RNAs for editing FLT3, CD123, and KIT are provided in the Table 1 below. As will be evident to one of ordinary skill in the art, selection of gRNA sequences can depend on factors such as the number of predicted on-target and/or off-target binding sites. In some embodiments, the gRNA sequence is selected to maximize potential on-target and minimize potential off-target sites.
In some embodiments, multiple gRNAs are introduced into the cell (e.g., one for FLT3 and one for CD123). In some embodiments, the two or more guide RNAs are transfected into cells in equimolar amounts. In some embodiments, the two or more guide RNAs are provided in amounts that are not equimolar. In some embodiments, the two or more guide RNAs are provided in amounts that are optimized so that editing of each target occurs at equal frequency. In some embodiments, the two or more guide RNAs are provided in amounts that are optimized so that editing of each target occurs at optimal frequency.
In some embodiments, provided herein is a “template” donor sequence. The template donor sequence includes a 100-500 nucleotide (e.g., 200 nucleotides) long single strand oligo-deoxynucleotide (ssODN), which functions as a donor template for homology directed repair (HDR), such that the desired mutation is introduced into the specific gene. Each donor template additionally includes selected silent mutations in bystander amino-acids to reduce the risk of re-cutting by the CRISPR-Cas ribonucleoprotein nuclease complex after successful DNA repair.
Exemplary template donor sequences for the introduction of the mutation N399D in FLT are provided in Table 2 below.
iv. Genetically Engineered Hematopoietic Cells
Provided herein are methods of producing the genetically engineered hematopoietic cells as described herein, which carry edited genes for expressing one or more cell-surface antigens in mutated form. Such methods can involve providing a cell and introducing into the cell components of a CRISPR Cas system for genome editing. In some embodiments, a nucleic acid that comprises a CRISPR-Cas guide RNA (gRNA) that hybridizes or is predicted to hybridize to a portion of the nucleotide sequence that encodes the cell-surface antigen is introduced into the cell. In some embodiments, the gRNA is introduced into the cell on a vector. In some embodiments, a Cas endonuclease is introduced into the cell. In some embodiments, the Cas endonuclease is introduced into the cell as a nucleic acid encoding a Cas endonuclease. In some embodiments, the gRNA and a nucleotide sequence encoding a Cas endonuclease are introduced into the cell on the same nucleic acid (e.g., the same vector). In some embodiments, the Cas endonuclease is introduced into the cell in the form of a protein. In some embodiments, the Cas endonuclease and the gRNA are pre-formed in vitro and are introduced to the cell in as a ribonucleoprotein complex.
v. Mutant FLT3
In some embodiments, the cell-surface protein is FLT3. The amino acid sequence of wild-type FLT3 is shown below:
In some embodiments, the methods described herein involve genetically engineering a population of hematopoietic cells using a Cas nuclease (or variant thereof). In some embodiments, the methods described herein involve genetically engineering a gene encoding a cell-surface antigen in a population of hematopoietic cells using a Cas nuclease or variant thereof (e.g., SpCas9 or AsCpf1). In some embodiments, the methods described herein involve genetically modifying or editing a FLT3 gene, or genetically modifying or editing a CD123 gene, or genetically modifying or editing a KIT gene, or genetically modifying or editing a FLT3 gene and a CD123 gene in the population of hematopoietic cells using the Cas nuclease.
In some embodiments, the methods described herein involve genetically engineering a mutant FLT3 gene in a population of hematopoietic cells using a Cas nuclease or variant thereof. In some embodiments, the methods described herein involve genetically engineering a mutation in exon 9 of FLT3 (e.g., thereby resulting in the mutation of position N399 in the encoded polypeptide) in a population of hematopoietic cells using a Cas nuclease or variant thereof. In some embodiments, the methods described herein involve genetically engineering a mutant FLT3 gene in a population of hematopoietic cells using a Cas nuclease or variant thereof and a guide sequence provided by any one of SEQ ID NOs: 13-23. In some embodiments, a template donor DNA sequence is also provided. For instance, a template donor DNA sequence provided by any one of SEQ ID NOs: 40-43.
In some embodiments, the genetically engineered FLT3 gene encodes a protein that has reduced binding to a therapeutic anti-FLT3 antibody. In some instances, the genetically engineered FLT3 gene includes at least one mutation in exon 9 of the FLT3 gene. In some instances, at least one mutation in exon 9 of the genetically engineered FLT3 gene results in a polypeptide bearing a mutation at position N399. In some instances, the mutation at position N399 is N399D or N399G. Exemplary amino acid sequences of the genetically engineered FLT3 are provided below:
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 51, wherein the polypeptide sequence comprises a mutation at N399D and wherein the polypeptide sequence has reduced binding to a therapeutic anti-FLT3 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 52, wherein the polypeptide sequence comprises a mutation at N399G and wherein the polypeptide sequence has reduced binding to a therapeutic anti-FLT3 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
vi. Mutant CD123
In some embodiments, the cell-surface protein is CD123. The amino acid sequence of wild-type CD123 is shown below:
In some embodiments, the methods described herein involve genetically engineering a mutant CD123 gene in a population of hematopoietic cells using a Cas nuclease or variant thereof. In some embodiments, the methods described herein involve genetically engineering a mutation in exon 2 of CD123 (e.g., thereby resulting in the mutation of position S59 in the encoded polypeptide) or in exon 3 of CD123 (e.g., thereby resulting in the mutation of position P88 in the encoded polypeptide) in a population of hematopoietic cells using a Cas nuclease or variant thereof. In some embodiments, the methods described herein involve genetically engineering a mutant CD123 gene in a population of hematopoietic cells using a Cas nuclease or variant thereof and a guide sequence provided by any one of SEQ ID NOs: 24-35.
In some embodiments, the genetically engineered HSPC includes a genetically engineered CD123 gene, wherein the genetically engineered CD123 gene encodes a protein that has reduced binding to a therapeutic anti-CD123 antibody. In some instances, the genetically engineered FLT3 gene includes at least one mutation in exon 2 or exon 3 of the CD123 gene. In some instances, at least one mutation in exon 2 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position S59. In some instances, the mutation at position S59 is S59P or S59F. In some instances, at least one mutation in exon 3 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position P88. In some instances, the mutation at position P88 is P88L or P88S. Exemplary amino acid sequences of the genetically engineered CD123 are provided below:
Nucleotide sequences are as follows:
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 54, wherein the polypeptide sequence comprises a mutation at S59P and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 55, wherein the polypeptide sequence comprises mutations at Y58H and S59P and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence. Also provided herein are nucleic acids encoding the polypeptide sequence.
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 56, wherein the polypeptide sequence comprises a mutation at S59F and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 57, wherein the polypeptide sequence comprises a mutation at P88S and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 58, wherein the polypeptide sequence comprises a mutation at P88L and wherein the polypeptide sequence has reduced binding to a therapeutic anti-CD123 antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
vii. Mutant KIT
In some embodiments, the cell-surface protein is KIT. The amino acid sequence of wild-type KIT is shown below:
In some embodiments, the methods described herein involve genetically engineering a mutant KIT gene in a population of hematopoietic cells using a Cas nuclease or variant thereof. In some embodiments, the methods described herein involve genetically engineering a mutation in exon 7 of KIT (e.g., mutating position H378) in a population of hematopoietic cells using a Cas nuclease or variant thereof. In some embodiments, the methods described herein involve genetically engineering a mutant KIT gene in a population of hematopoietic cells using a Cas nuclease or variant thereof and a guide sequence provided by any one of SEQ ID NOs: 36-47.
In some embodiments, the genetically engineered HSPC includes a genetically engineered KIT gene, wherein the genetically engineered KIT gene encodes a protein that has reduced binding to a therapeutic anti-KIT antibody. In some instances, the genetically engineered KIT gene includes at least one mutation in exon 6 and/or exon 7 of the KIT gene. In some instances, at least one mutation in exon 7 of the genetically engineered KIT gene results in a polypeptide bearing a mutation at position H378. In some instances, the mutation at position H378 is H378R or H378S or H378P or H378A or H378F or H378K or H378G or H378L or H378M. In some instances, mutation in exon 6 of KIT results in the encoded polypeptide having one or more of the following mutations F316S, M318V, I319K, V323I, 1334V, E360K, P363V, E366D. In some instances, the KIT exon 7 mutation results in a polypeptide having mutations at E376Q and/or H378R.
Exemplary amino acid sequences of the genetically engineered KIT are provided below:
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 67, wherein the polypeptide sequence comprises mutations at F316S, M318V, I319K, V323I, 1334V, E360K, P363V, E366D, E376Q, and H378R and wherein the polypeptide sequence has reduced binding to a therapeutic anti-KIT antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
In some embodiments, provided herein is a polypeptide sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence set forth in SEQ ID NO: 68, wherein the polypeptide sequence comprises a mutation at H378R and wherein the polypeptide sequence has reduced binding to a therapeutic anti-KIT antibody. Also provided herein are nucleic acids encoding the polypeptide sequence.
viii. Genetically Engineered Hematopoietic Cells Expressing Mutant FLT3, CD123, and or KIT Genes
In some embodiments, the cell-surface proteins are FLT3 and CD123. In some embodiments, the cell-surface proteins are FLT3 and KIT. In some embodiments, the cell-surface proteins are KIT and CD123. In some embodiments, the two or more guides are transfected concurrently with each other. In some embodiments, the two or more guides are provided sequentially or consecutively, i.e., in two or more separate transfections. For example, a FLT3 guide RNA (any one of SEQ ID NOs: 13-23), a CD123 guide RNA (any one of SEQ ID NOs: 24-35), a KIT guide RNA (any one of SEQ ID NOs: 36-47).
In some embodiments, the genetically engineered HSPC includes a genetically engineered FLT3 gene, wherein the genetically engineered FLT3 gene encodes a protein that has reduced binding to a therapeutic anti-FLT3 antibody. In some instances, the genetically engineered FLT3 gene includes at least one mutation in exon 9 of the FLT3 gene. In some instances, at least one mutation in exon 9 of the genetically engineered FLT3 gene results in a polypeptide bearing a mutation at position N399. In some instances, the mutation at position N399 is N399D or N399G. Exemplary amino acid sequences of the genetically engineered FLT3 are shown above (see SEQ ID NOs: 49-52).
In some embodiments, the genetically engineered HSPC includes a genetically engineered CD123 gene, wherein the genetically engineered CD123 gene encodes a protein that has reduced binding to a therapeutic anti-CD123 antibody. In some instances, the genetically engineered CD123 gene includes at least one mutation in exon 2 or exon 3 of the CD123 gene. In some instances, a mutation in exon 2 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position S59. In some instances, the mutation at position S59 is S59P or S59F. In some instances, the mutations are S59P and Y58H. In some instances, at least one mutation in exon 3 of the genetically engineered CD123 gene results in a polypeptide bearing a mutation at position P88. In some instances, the mutation at position P88 is P88L or P88S. Exemplary amino acid sequences of the genetically engineered CD123 are shown above (see SEQ ID NOs: 54-58).
In some embodiments, the genetically engineered HSPC includes a genetically engineered KIT gene, wherein the genetically engineered KIT gene encodes a protein that has reduced binding to a therapeutic anti-KIT antibody. In some instances, the genetically engineered KIT gene includes at least one mutation in exon 6 and/or exon 7 of the KIT gene. In some instances, at least one mutation in exon 7 of the genetically engineered KIT gene results in a polypeptide bearing a mutation at position H378. In some instances, the mutation at position H378 is H378R or H378S or H378P or H378A or H378F or H378K or H378G or H378L or H378M. In some instances, the mutations in exon 6 of KIT results in the encoded polypeptide having one or more of the following mutations: F316S, M318V, I319K, V323I, 1334V, E360K, P363V, E366D. In some instances, the mutations of exon 7 in KIT result in a polypeptide having mutations at one or more of E376Q and H378R. Exemplary amino acid sequences of the genetically engineered KIT are provided are shown above (see SEQ ID NOs: 67-68).
Cytotoxic agents targeting cells (e.g., cancer cells) expressing a cell-surface antigen can be co-used with the genetically engineered hematopoietic cells as described herein. As used herein, the term “cytotoxic agent” refers to any agent that can directly or indirectly induce cytotoxicity of a target cell, which expresses the specific cell-surface antigen (e.g., a target cancer cell). Such a cytotoxic agent can comprise a protein-binding fragment that binds and targets an epitope of the specific cell-surface antigen.
i. Therapeutic Antibodies Antibody-Drug Conjugates
In some instances, the cytotoxic agent includes a therapeutic antibody, which can be conjugated to a drug (e.g., an anti-cancer drug) to form an antibody-drug conjugate (ADC). In some embodiments, the agent is an antibody-drug conjugate. In some embodiments, the antibody-drug conjugate comprises an epitope binding fragment and a toxin or drug that induces cytotoxicity in a target cell.
In some embodiments, the therapeutic anti-FLT3 antibody is anti-FLT3 clone 4G8 antibody. In some embodiments, the therapeutic anti-CD123 antibody is clone 7G3 antibody or its humanized counterpart CSL362 (“talacotuzumab”). In some embodiments, the therapeutic anti-CD123 antibody is anti-CD123 clone 6H6 antibody or anti-CD123 clone S18016F antibody. In some embodiments, the therapeutic anti-KIT antibody is anti-KIT clone Fab79D antibody.
Toxins or drugs compatible for use in antibody-drug conjugates are well known in the art and will be evident to one of ordinary skill in the art. See, e.g., Peters et al. Biosci. Rep. (2015) 35 (4): e00225, Beck et al. Nature Reviews Drug Discovery (2017) 16:315-337; Marin-Acevedo et al. J. Hematol. Oncol. (2018) 11:8; Elgundi et al. Advanced Drug Delivery Reviews (2017) 122:2-19. In some embodiments, the antibody-drug conjugate can further comprise a linker (e.g., a peptide linker, such as a cleavable linker or a non-cleavable linker) attaching the antibody and drug molecule. Examples of antibody-drug conjugates include, without limitation, brentuximab vedotin, glembatumumab vedotin/CDX-011, depatuxizumab mafodotin/ABT-414, PSMA ADC, polatuzumab vedotin/RG7596/DCDS4501A, denintuzumab mafodotin/SGN-CD19A, AGS-16C3F, CDX-014, RG7841/DLYE5953A, RG7882/DMUC406A, RG7986/DCDS0780A, SGN-LIV1A, enfortumab vedotin/ASG-22ME, AG-15ME, AGS67E, telisotuzumab vedotin/ABBV-399, ABBV-221, ABBV-085, GSK-2857916, tisotumab vedotin/HuMax-TF-ADC, HuMax-Axl-ADC, pinatuzumab veodtin/RG7593/DCDT2980S, lifastuzumab vedotin/RG7599/DNIB0600A, indusatumab vedotin/MLN-0264/TAK-264, vandortuzumab vedotin/RG7450/DSTP3086S, sofituzumab vedotin/RG7458/DMUC5754A, RG7600/DMOT4039A, RG7336/DEDN6526A, ME1547, PF-06263507/ADC 5T4, trastuzumab emtansine/T-DM1, mirvetuximab soravtansine/IMGN853, coltuximab ravtansine/SAR3419, naratuximab emtansine/IMGN529, indatuximab ravtansine/BT-062, anetumab ravtansine/BAY 94-9343, SAR408701, SAR428926, AMG 224, PCA062, HKT288, LY3076226, SAR566658, lorvotuzumab mertansine/IMGN901, cantuzumab mertansine/SB-408075, cantuzumab ravtansine/IMGN242, laprituximab emtansine/IMGN289, IMGN388, bivatuzumab mertansine, AVE9633, BIIB015, MLN2704, AMG 172, AMG 595, LOP 628, vadastuximab talirine/SGN-CD33A, SGN-CD70A, SGN-CD19B, SGN-CD123A, SGN-CD352A, rovalpituzumab tesirine/SC16LD6.5, SC-002, SC-003, ADCT-301/HuMax-TAC-PBD, ADCT-402, MEDI3726/ADC-401, IMGN779, IMGN632, gemtuzumab ozogamicin, inotuzumab ozogamicin/CMC-544, PF-06647263, CMD-193, CMB-401, trastuzumab duocarmazine/SYD985, BMS-936561/MDX-1203, sacituzumab govitecan/IMMU-132, labetuzumab govitecan/IMMU-130, DS-8201a, U3-1402, milatuzumab doxorubicin/IMMU-110/hLL1-DOX, BMS-986148, RC48-ADC/hertuzumab-vc-MMAE, PF-06647020, PF-06650808, PF-06664178/RN927C, lupartumab amadotin/BAY1129980, aprutumab ixadotin/BAY1187982, ARX788, AGS62P1, XMT-1522, AbGn-107, MEDI4276, DSTA4637S/RG7861. In one example, the antibody-drug conjugate is gemtuzumab ozogamicin.
In some embodiments, binding of the antibody-drug conjugate to the epitope of the cell-surface protein induces internalization of the antibody-drug conjugate, and the drug (or toxin) can be released intracellularly. In some embodiments, binding of the antibody-drug conjugate to the epitope of a cell-surface protein induces internalization of the toxin or drug, which allows the toxin or drug to kill the cells expressing the cell surface protein (target cells). In some embodiments, binding of the antibody-drug conjugate to the epitope of a cell-surface protein induces internalization of the toxin or drug, which can regulate the activity of the cell expressing the cell surface protein (target cells). The type of toxin or drug used in the antibody-drug conjugates described herein is not limited to any specific type.
In some embodiments, two or more (e.g., 2, 3, 4, 5 or more) epitopes of a cell-surface antigen have been modified, enabling two or more (e.g., 2, 3, 4, 5 or more) different cytotoxic agents (e.g., two ADCs) to be targeted to the two or more epitopes. In some embodiments, the toxins carried by the ADCs could work synergistically to enhance efficacy (e.g., death of the target cells). In some embodiments, epitopes of two or more (e.g., 2, 3, 4, 5 or more) cell-surface proteins have been modified, enabling two or more (e.g., 2, 3, 4, 5 or more) different cytotoxic agents (e.g., two ADCs) to be targeted to epitopes of the two or more cell-surface antigens. In some embodiments, one or more (e.g., 1, 2, 3, 4, 5 or more) epitopes of a cell-surface antigen have been modified and one or more (e.g., 1, 2, 3, 4, 5 or more) epitopes of an additional cell-surface protein have been modified, enabling two or more (e.g., 2, 3, 4, 5 or more) different cytotoxic agents (e.g., two ADCs) to be targeted to epitopes of the cell-surface antigen and epitopes of additional cell-surface antigen. In some embodiments, targeting of more than one cell-surface antigen or a cell-surface antigen and one or more additional cell-surface protein/antigen can reduce relapse of a hematopoietic malignancy.
In some embodiments, the methods described herein involve administering ADCs that target an epitope of a cell-surface antigen that is mutated in the population of genetically engineered hematopoietic cells. In some embodiments, the methods described herein involve administering ADCs that target an epitope of a cell-surface antigen that is mutated in the population of genetically engineered hematopoietic cells and one or more additional cytotoxic agents that can target one or more additional cell-surface proteins. In some embodiments, the agents could work synergistically to enhance efficacy by targeting more than one cell-surface protein.
An ADC described herein can be used as a follow-on treatment to subjects who have been undergone the combined therapy as described herein.
In some embodiments, the methods described herein involve administering to the subject a population of genetically engineered cells lacking a non-essential epitope in a cell-surface antigen and one or more immunotherapeutic agents (e.g., ADCs) that target cells expressing the cell-surface antigen. In some embodiments, the methods described herein involve administering to the subject a population of genetically engineered cells lacking a non-essential epitope in a type 1 cell-surface antigen and one or more immunotherapeutic agents (e.g., ADCs) that target cells expressing the cell-surface antigen. In some embodiments, the methods described herein involve administering to the subject a population of genetically engineered cells lacking a non-essential epitope in a type 2 cell-surface antigen and one or more immunotherapeutic agents (e.g., ADCs) that target cells expressing the cell-surface antigen. In any of the embodiments described herein, one or more additional immunotherapeutic agents can be further administered to the subject (e.g., targeting one or more additional epitopes and/or antigens), for example if the hematopoietic malignancy relapses.
ii. Immune Cells Expressing Chimeric Antigen Receptors
In some embodiments, the cytotoxic agent that targets an epitope of a specific cell-surface antigen as described herein is an immune cell that expresses a chimeric receptor, which comprises an epitope binding fragment (e.g., a single-chain antibody) capable of binding to the epitope of the cell surface protein (e.g., FLT3, CD123, or KIT). Recognition of a target cell (e.g., a cancer cell) having the epitope of the specific protein on its cell surface by the epitope binding fragment of the chimeric receptor transduces an activation signal to the signaling domain(s) (e.g., co-stimulatory signaling domain and/or the cytoplasmic signaling domain) of the chimeric receptor, which can activate an effector function in the immune cell expressing the chimeric receptor.
In some embodiments, the immune cell expresses more than one chimeric receptor (e.g., 2, 3, 4, 5 or more), referred to as a bispecific or multi-specific immune cell. In some embodiments, the immune cell expresses more than one chimeric receptor, at least one of which targets an epitope of a cell-surface antigen. In some embodiments, the immune cell expresses more than one chimeric receptor, each of which targets an epitope of a specific cell-surface antigen. In some embodiments, the immune cell expresses more than one chimeric receptor, at least one of which targets an epitope of a cell-surface antigen and at least one of which targets an epitope of an additional cell-surface antigen. In some embodiments, targeting of more than one cell-surface protein or a cell-surface protein and one or more additional cell-surface protein can reduce relapse of a hematopoietic malignancy. In some embodiments, the immune cell expresses a chimeric receptor that targets more than one epitope (e.g., more than one epitope of one antigen or epitopes of more than one antigen), referred to as a bispecific chimeric receptor.
In some embodiments, epitopes of two or more lineage-specific cell-surface proteins are targeted by cytotoxic agents. In some embodiments, two or more chimeric receptors are expressed in the same immune cell, e.g., bispecific chimeric receptors. Such cells can be used in any of the methods described herein. In some embodiments, cells expressing a chimeric receptor are “pooled”, i.e., two or more groups of cells express two or more different chimeric receptors. In some embodiments, two or more cells expressing different chimeric antigen receptors are administered concurrently. In some embodiments, two or more cells expressing different chimeric antigen receptors are administered sequentially. In some embodiments, epitopes of FLT3, CD123, and/or KIT are targeted by cytotoxic agents. In some embodiments, the chimeric receptors targeting FLT3, CD123, and/or KIT are expressed in the same immune cell (i.e., a bispecific immune cell). Such cells can be used in any of the methods described herein. In some embodiments, cells expressing chimeric receptors targeting FLT3, CD123, and/or KIT “pooled”, i.e., two or more groups of cells express two or more different chimeric receptors. In some embodiments, two or more groups of cells expressing chimeric receptors targeting FLT3, CD123, and/or KIT are administered concurrently. In some embodiments, two or more groups of cells expressing chimeric receptors targeting FLT3, CD123, and/or KIT are administered sequentially.
As used herein, a chimeric receptor refers to a non-naturally occurring molecule that can be expressed on the surface of a host cell and comprises binding domain that provides specificity of the chimeric receptor (e.g., an epitope binding fragment that binds to an epitope of a cell-surface lineage-specific protein). In general, chimeric receptors comprise at least two domains that are derived from different molecules. In addition to the epitope-binding fragment described herein, the chimeric receptor may further comprise one or more of the following: a hinge domain (e.g., CD28 hinge, IgG4 hinge, or CD8α hinge), a transmembrane domain (e.g., CD28 TM, CD8α TM, 4-1BB TM), a co-stimulatory domain (e.g., CD28z, 4-1BB, ICOS, OX40), a cytoplasmic signaling domain (e.g., CD3z), and combinations thereof.
In some embodiments, the chimeric receptors described herein comprise one or more hinge domain(s). In some embodiments, the hinge domain may be located between the epitope binding fragment and a transmembrane domain. A hinge domain is an amino acid segment that is generally found between two domains of a protein and may allow for flexibility of the protein and movement of one or both of the domains relative to one another. Any amino acid sequence that provides such flexibility and movement of the epitope binding fragment relative to another domain of the chimeric receptor can be used.
The hinge domain may contain about 10-200 amino acids, e.g., 15-150 amino acids, 20-100 amino acids, or 30-60 amino acids. In some embodiments, the hinge domain may be of about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 amino acids in length.
In some embodiments, the hinge domain is a hinge domain of a naturally occurring protein. Hinge domains of any protein known in the art to comprise a hinge domain are compatible for use in the chimeric receptors described herein. In some embodiments, the hinge domain is at least a portion of a hinge domain of a naturally occurring protein and confers flexibility to the chimeric receptor. In some embodiments, the hinge domain is of CD8α or CD28. In some embodiments, the hinge domain is a portion of the hinge domain of CD8a, e.g., a fragment containing at least 15 (e.g., 20, 25, 30, 35, or 40) consecutive amino acids of the hinge domain of CD8α or CD28.
Hinge domains of antibodies, such as an IgG, IgA, IgM, IgE, or IgD antibody, are also compatible for use in the chimeric receptors described herein. In some embodiments, the hinge domain is the hinge domain that joins the constant domains CH1 and CH2 of an antibody. In some embodiments, the hinge domain is of an antibody and comprises the hinge domain of the antibody and one or more constant regions of the antibody. In some embodiments, the hinge domain comprises the hinge domain of an antibody and the CH3 constant region of the antibody. In some embodiments, the hinge domain comprises the hinge domain of an antibody and the CH2 and CH3 constant regions of the antibody. In some embodiments, the antibody is an IgG, IgA, IgM, IgE, or IgD antibody. In some embodiments, the antibody is an IgG antibody. In some embodiments, the antibody is an IgG1, IgG2, IgG3, or IgG4 antibody. In some embodiments, the hinge region comprises the hinge region and the CH2 and CH3 constant regions of an IgG1 antibody. In some embodiments, the hinge region comprises the hinge region and the CH3 constant region of an IgG1 antibody.
In some embodiments, the chimeric receptors described herein may comprise one or more transmembrane domain(s). The transmembrane domain for use in the chimeric receptors can be in any form known in the art. As used herein, a “transmembrane domain” refers to any protein structure that is thermodynamically stable in a cell membrane, preferably a eukaryotic cell membrane. Transmembrane domains compatible for use in the chimeric receptors used herein may be obtained from a naturally occurring protein. Alternatively, the transmembrane domain may be a synthetic, non-naturally occurring protein segment, e.g., a hydrophobic protein segment that is thermodynamically stable in a cell membrane.
Transmembrane domains are classified based on the transmembrane domain topology, including the number of passes that the transmembrane domain makes across the membrane and the orientation of the protein. For example, single-pass membrane proteins cross the cell membrane once, and multi-pass membrane proteins cross the cell membrane at least twice (e.g., 2, 3, 4, 5, 6, 7 or more times). In some embodiments, the transmembrane domain is a single-pass transmembrane domain. In some embodiments, the transmembrane domain is a single-pass transmembrane domain that orients the N terminus of the chimeric receptor to the extracellular side of the cell and the C terminus of the chimeric receptor to the intracellular side of the cell. In some embodiments, the transmembrane domain is obtained from a single pass transmembrane protein. In some embodiments, the transmembrane domain is of CD28 or 4-1BB or CD8a.
In some embodiments, the chimeric receptors described herein comprise one or more costimulatory signaling domains. The term “co-stimulatory signaling domain,” as used herein, refers to at least a portion of a protein that mediates signal transduction within a cell to induce an immune response, such as an effector function. The co-stimulatory signaling domain of the chimeric receptor described herein can be a cytoplasmic signaling domain from a co-stimulatory protein, which transduces a signal and modulates responses mediated by immune cells, such as T cells, NK cells, macrophages, neutrophils, or eosinophils.
In some embodiments, the chimeric receptor comprises more than one (at least 2, 3, 4, or more) co-stimulatory signaling domains. In some embodiments, the chimeric receptor comprises more than one co-stimulatory signaling domains obtained from different costimulatory proteins. In some embodiments, the chimeric receptor does not comprise a co-stimulatory signaling domain.
In general, many immune cells require co-stimulation, in addition to stimulation of an antigen-specific signal, to promote cell proliferation, differentiation and survival, and to activate effector functions of the cell. Activation of a co-stimulatory signaling domain in a host cell (e.g., an immune cell) may induce the cell to increase or decrease the production and secretion of cytokines, phagocytic properties, proliferation, differentiation, survival, and/or cytotoxicity. The co-stimulatory signaling domain of any co-stimulatory protein may be compatible for use in the chimeric receptors described herein. The type(s) of co-stimulatory signaling domain is selected based on factors such as the type of the immune cells in which the chimeric receptors would be expressed (e.g., primary T cells, T cell lines, NK cell lines) and the desired immune effector function (e.g., cytotoxicity). Examples of co-stimulatory signaling domains for use in the chimeric receptors can be the cytoplasmic signaling domain of co-stimulatory proteins, including, without limitation, CD27, CD28ζ (CD28z), 4-1BB, OX40, CD30, ICOS, CD2, CD7, LIGHT, NKG2C, B7-H3.
In some embodiments, the chimeric receptors described herein comprise one or more cytoplasmic signaling domain(s). Any cytoplasmic signaling domain can be used in the chimeric receptors described herein. In general, a cytoplasmic signaling domain relays a signal, such as interaction of an extracellular ligand-binding domain with its ligand, to stimulate a cellular response, such as inducing an effector function of the cell (e.g., cytotoxicity). In some embodiments, the cytoplasmic signaling domain is from CD3ζ (CD3z).
In some embodiments, provided herein a chimeric receptor construct targeting FLT3, CD123, FLT3+CD123, KIT, FLT3+KIT or KIT+CD123. The construct can further include at least a hinge domain (e.g., from CD28, CD8α, or an antibody), a transmembrane domain (e.g., from CD28), one or more co-stimulatory domains (from one or more of CD28z) and a cytoplasmic signaling domain (e.g., from CD3z), or a combination thereof. In some examples, the methods described herein involve administering to a subject a population of genetically engineered hematopoietic cells (engineered to have a mutant FLT3, CD123, FLT3+CD123, KIT, FLT3+KIT or CD123+KIT) and an immune cell expressing a chimeric receptor that targets FLT3, CD123, FLT3+CD123, KIT, FLT3+KIT or CD123+KIT respectively, which may further comprise at least a hinge domain (e.g., from CD28, CD8α, or an antibody), a transmembrane domain (e.g., from CD28), one or more co-stimulatory domains (from one or more of CD28z) and a cytoplasmic signaling domain (e.g., from CD3z), or combination thereof. In some embodiments, the administered immunotherapeutic product is a combination of immune cells expressing individual chimeric receptor that targets FLT3, CD123, and/or KIT.
Any of the chimeric receptors described herein can be prepared by routine methods, such as recombinant technology. Methods for preparing the chimeric receptors herein involve generation of a nucleic acid that encodes a polypeptide comprising each of the domains of the chimeric receptors, including the epitope binding fragment and optionally, the hinge domain, the transmembrane domain, at least one co-stimulatory signaling domain, and the cytoplasmic signaling domain. In some embodiments, nucleic acids encoding the components of a chimeric receptor are joined together using recombinant technology.
Additionally, any of the chimeric receptors can be expressed in immune cells and administered to a subject (e.g., a human subject) by routine methods. For example, T cells can be either derived from T cells in a subject's own blood (autologous) or derived from the T cells of another healthy donor (allogeneic). Once isolated from a subject, these T cells are genetically engineered to express a specific CAR, which programs them to target an antigen that is present on the surface of tumors. The CAR-T cells are then infused, by customary practice, into the subject.
In some embodiments, the CAR is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of any one of SEQ ID NOs: 69, 71, 73, 75, 77, 79, 86, or 87, wherein the CAR retains its ability to bind to its respective cell-surface lineage-specific protein (e.g., KIT, CD123, FLT3, or a combination thereof).
In some embodiments, the cell-surface lineage-specific protein is KIT and the epitope binding fragment comprises the following CDR sequences: GFNISVYMMH (SEQ ID NO: 88), SIYPYSGYTYYADSVKG (SEQ ID NO: 89), ARYVYHALDY (SEQ ID NO: 90), RASQRGLRNVAVA (SEQ ID NO: 91), SASSLYS (SEQ ID NO: 92), and QQWAVHSLIT (SEQ ID NO: 93). In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to GFNISVYMMHWVRQAPGKGLEWVASIYPYSGYTYYADSVKGRFTISADTSKNT AYLQMNSLRAEDTAVYYCARYVYHALDY (SEQ ID NO: 94), wherein the epitope binding fragment retains its ability to bind to its respective KIT epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to RASQRGLRNVAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTDFTLTIS SLQPEDFATYYCQQWAVHSLIT (SEQ ID NO: 95), wherein the epitope binding fragment retains its ability to bind to its respective KIT epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 94 and comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 95. In some embodiments, the epitope binding fragment comprises both SEQ ID NOs 94 and 95.
In some embodiments, the cell-surface lineage-specific protein is FLT3 and the epitope binding fragment comprises the following CDR sequences: GYTFTSYWMH (SEQ ID NO: 96), EIDPSDSYKDYNQKFK (SEQ ID NO: 97), RAITTTPFDF (SEQ ID NO: 98), RASQSISNNLH (SEQ ID NO: 99), YASQSIS (SEQ ID NO: 100), and QQSNTWPYT (SEQ ID NO: 101). In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to GYTFTSYWMHWVRQRPGHGLEWIGEIDPSDSYKDYNQKFKDKATLTVDRSSNT AYMHLSSLTSDDSAVYYCARAITTTPFDF (SEQ ID NO: 102), wherein the epitope binding fragment retains its ability to bind to its respective FLT3 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to RASQSISNNLHWYQQKSHESPRLLIKYASQSISGIPSRFSGSGSGTDFTLSINSVETE DFGVYFCQQSNTWPYT (SEQ ID NO: 103), wherein the epitope binding fragment retains its ability to bind to its respective FLT3 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 102 and comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 103. In some embodiments, the epitope binding fragment comprises both SEQ ID NOs 102 and 103.
In some embodiments, the cell-surface lineage-specific protein is CD123 and the epitope binding fragment comprises the following CDR sequences: GYSFTDYYMK (SEQ ID NO: 104), DIIPSNGATFYNQKFKG (SEQ ID NO: 105), ARSHLLRASWFAY (SEQ ID NO: 106), SQSLLNSGNQKNYLT (SEQ ID NO: 107), WASTRES (SEQ ID NO: 108), and QNDYSYPYT (SEQ ID NO: 109). In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to GYSFTDYYMKWARQMPGKGLEWMGDIIPSNGATFYNQKFKGQVTISADKSISTT YLQWSSLKASDTAMYYCARSHLLRASWFAY (SEQ ID NO: 110), wherein the epitope binding fragment retains its ability to bind to its respective CD123 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to ESSQSLLNSGNQKNYLTWYQQKPGQPPKPLIYWASTRESGVPDRFSGSGSGTDFT LTISSLQAEDVAVYYCONDYSYPYT (SEQ ID NO: 111), wherein the epitope binding fragment retains its ability to bind to its respective CD123 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 110 and comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 111. In some embodiments, the epitope binding fragment comprises both SEQ ID NOs 110 and 111.
In some embodiments, the cell-surface lineage-specific proteins are FLT3 and CD123 and the epitope binding fragment comprises the following CDR sequences: GYTFTSYWMH (SEQ ID NO: 96), EIDPSDSYKDYNQKFK (SEQ ID NO: 97), RAITTTPFDF (SEQ ID NO: 98), RASQSISNNLH (SEQ ID NO: 99), YASQSIS (SEQ ID NO: 100), QQSNTWPYT (SEQ ID NO: 101), GYSFTDYYMK (SEQ ID NO: 104), DIIPSNGATFYNQKFKG (SEQ ID NO: 105), ARSHLLRASWFAY (SEQ ID NO: 106), SQSLLNSGNQKNYLT (SEQ ID NO: 107), WASTRES (SEQ ID NO: 108), and QNDYSYPYT (SEQ ID NO: 109). In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to GYTFTSYWMHWVRQRPGHGLEWIGEIDPSDSYKDYNQKFKDKATLTVDRSSNT AYMHLSSLTSDDSAVYYCARAITTTPFDF (SEQ ID NO: 102), wherein the epitope binding fragment retains its ability to bind to its respective FLT3 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to RASQSISNNLHWYQQKSHESPRLLIKYASQSISGIPSRFSGSGSGTDFTLSINSVETE DFGVYFCQQSNTWPYT (SEQ ID NO: 103), wherein the epitope binding fragment retains its ability to bind to its respective FLT3 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to GYSFTDYYMKWARQMPGKGLEWMGDIIPSNGATFYNQKFKGQVTISADKSISTT YLQWSSLKASDTAMYYCARSHLLRASWFAY (SEQ ID NO: 110), wherein the epitope binding fragment retains its ability to bind to its respective CD123 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to ESSQSLLNSGNQKNYLTWYQQKPGQPPKPLIYWASTRESGVPDRFSGSGSGTDFT LTISSLQAEDVAVYYCONDYSYPYT (SEQ ID NO: 111), wherein the epitope binding fragment retains its ability to bind to its respective CD123 epitope. In some instances, the epitope binding fragment comprises a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 102, a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 103, a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 110, and a sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 111. In some embodiments, the epitope binding fragment comprises all four of SEQ ID NOs 102, 103, 110, and 111.
In some embodiments, the cell-surface lineage-specific protein is CD123 and the epitope binding fragment comprises the following CDR sequences: DIIPSNGATFYNQKFKG (SEQ ID NO: 105), SQSLLNSGNQKNYLT (SEQ ID NO: 107), WASTRES (SEQ ID NO: 108), and QNDYSYPYT (SEQ ID NO: 109).
RQAPGKGLEWVA
SIYPYSGYTYYADSVKG
RFTISADTSKNTAYLQMNSLRAEDTAVYYC
ARYVYHALDY
WGQGTLVTVSSGSTSGSGKPGSSEGSTKGDIQMTQSPSSLSASVGDRVT
LQPEDFATYYC
QQWAVHSLIT
FGQGTKVEIKRAAIEVMYPPPYLDNEKSNGTIIHVKGK
QQKPGKAPKLLIY
SASSLYS
GVPSRFSGSRSGTDFTLTISSLQPEDFATYYC
QQWAVHS
LIT
FGQGTKVEIKGSTSGSGKPGSGEGSTKGEVQLVESGGGLVQPGGSLRLSCAASGFN
ISVYMMH
WVRQAPGKGLEWVA
SIYPYSGYTYYADSVKG
RFTISADTSKNTAYLQMNSLR
AEDTAVYYC
ARYVYHALDY
WGQGTLVTVSSIEVMYPPPYLDNEKSNGTIIHVKGKHLCP
RQRPGHGLEWIG
EIDPSDSYKDYNQKFK
DKATLTVDRSSNTAYMHLSSLTSDDSAVYYC
A
RAITTTPFDF
WGQGTTLTVSSGGGGSGGGGSGGGGSDIVLTQSPATLSVTPGDSVSLS
DFGVYFC
QQSNTWPYT
FGGGTKLEIKRESKYGPPCPPCPASMFWVLVVVGGVLACYSLL
RQMPGKGLEWMG
DIIPSNGATFYNQKFKG
QVTISADKSISTTYLQWSSLKASDTAMYYC
ARSHLLRASWFAY
WGQGTMVTVSSGSTSGSGKPGSSEGSTKGDIVMTQSPDSLAVSLGE
FTLTISSLQAEDVAVYYC
QNDYSYPYT
FGQGTKLEIKTTTPAPRPPTPAPTIASQPLSL
RQRPGHGLEWIG
EIDPSDSYKDYNQKFK
DKATLTVDRSSNTAYMHLSSLTSDDSAVYYC
A
RAITTTPFDF
WGQGTTLTVSSGSTSGSGKPGSSEGSTKGDIVLTQSPATLSVTPGDSV
VPDRFSGSGSGTDFTLTISSLQAEDVAVYYC
QNDYSYPYT
FGQGTKLEIKSGSTSGSGK
DIIPSNGATFYNQKFKGQVTISADKSISTTYLQWSSLKASDTAMYYCARSHLLRASWFA
Y
WGQGTMVTVSTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDMFW
QKSHESPRLLIK
YASQSIS
GIPSRFSGSGSGTDFTLSINSVETEDFGVYFC
QQSNTWPY
T
FGGGTKLEIKRGSTSGSGKPGSSEGSTKGQVQLQQPGAELVKPGASLKLSCKSSGYTF
TSYWMHWVRQRPGHGLEWIGEIDPSDSYKDYNQKFKDKATLTVDRSSNTAYMHLSSLTS
FKG
QVTISADKSISTTYLQWSSLKASDTAMYYC
ARSHLLRASWFAY
WGQGTMVTVSSGS
QKPGQPPKPLIY
WASTRES
GVPDRFSGSGSGTDFTLTISSLQAEDVAVYYC
QNDYSYPY
T
FGQGTKLEIKTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDMFW
The genetically engineered hematopoietic cells such as HSCs can be administered to a subject in need of the treatment, either taken alone or in combination of one or more cytotoxic agents that target one or more cell-surface antigens as described herein. Since the hematopoietic cells are genetically edited in the genes of the one or more cell-surface antigens, the hematopoietic cells and/or descendant cells thereof would express the one or more cell-surface antigens in mutated form (e.g., but functional) such that they can escape being targeted by the cytotoxic agents, for example, CAR-T cells.
Thus, the present disclosure provides methods for treating a hematopoietic malignancy, the method comprising administering to a subject in need thereof (i) a population of the genetically engineered hematopoietic cells described herein, and optionally (ii) a cytotoxic agent such as CAR-T cells that target a cell-surface antigen, the gene of which is genetically edited in the hematopoietic cells such that the cytotoxic agent does not target hematopoietic cells or descendant cells thereof. The administration of (i) and (ii) can be concurrently or in any order. In some embodiments, the cytotoxic agents and/or the hematopoietic cells can be mixed with a pharmaceutically acceptable carrier to form a pharmaceutical composition, which is also within the scope of the present disclosure.
As used herein, “subject,” “individual,” and “patient” are used interchangeably, and refer to a vertebrate, preferably a mammal such as a human. Mammals include, but are not limited to, human primates, non-human primates or murine, bovine, equine, canine or feline species. In some embodiments, the subject is a human patient having a hematopoietic malignancy.
To perform the methods described herein, an effective amount of the genetically engineered hematopoietic cells can be administered to a subject in need of the treatment. Optionally, the hematopoietic cells can be co-used with a cytotoxic agent as described herein. As used herein the term “effective amount” can be used interchangeably with the term “therapeutically effective amount” and refers to that quantity of a cytotoxic agent, hematopoietic cell population, or pharmaceutical composition (e.g., a composition comprising cytotoxic agents and/or hematopoietic cells) that is sufficient to result in a desired activity upon administration to a subject in need thereof. Within the context of the present disclosure, the term “effective amount” refers to that quantity of a compound, cell population, or pharmaceutical composition that is sufficient to delay the manifestation, arrest the progression, relieve or alleviate at least one symptom of a disorder treated by the methods of the present disclosure. Note that when a combination of active ingredients is administered the effective amount of the combination can or cannot include amounts of each ingredient that would have been effective if administered individually.
Effective amounts vary, as recognized by those skilled in the art, depending on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size, gender and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. In some embodiments, the effective amount alleviates, relieves, ameliorates, improves, reduces the symptoms, or delays the progression of any disease or disorder in the subject. In some embodiments, the subject is a human. In some embodiments, the subject is a human patient having a hematopoietic malignancy.
As described herein, the hematopoietic cells and/or immune cells expressing chimeric receptors can be autologous to the subject, i.e., the cells are obtained from the subject in need of the treatment, manipulated such that the cells do not bind the cytotoxic agents, and then administered to the same subject. Administration of autologous cells to a subject can result in reduced rejection of the host cells as compared to administration of non-autologous cells. For example, HSPCs are obtained from a biological sample from a subject, the HSPCs are genetically engineered, and the genetically engineered HSPCs are administered to the same subject. In some instances, the HSPCs are obtained from a biological sample, wherein the biological sample is bone marrow cells, blood, cord blood cells, or mobilized peripheral blood-derived CD34+ hematopoietic stem and progenitor cells.
Alternatively, the host cells are allogeneic cells, i.e., the cells are obtained from a first subject, genetically engineered, and then administered to a second subject that is different from the first subject but of the same species. For example, allogeneic immune cells can be derived from a human donor and administered to a human recipient who is different from the donor. In some embodiments, the hematopoietic cells have been further genetically engineered to reduce host-versus-graft effects. For example, in some embodiments, immune cells and/or hematopoietic cells can be subjected to gene editing or silencing methods to reduce or eliminate expression of one or more proteins involved in inducing host immune responses.
A typical amount of cells, i.e., immune cells or hematopoietic cells, administered to a mammal (e.g., a human) can be, for example, in the range of about 106 to 1011 cells. In some embodiments it can be desirable to administer fewer than 106 cells to the subject. In some embodiments, it can be desirable to administer more than 1011 cells to the subject. In some embodiments, one or more doses of cells includes about 106 cells to about 1011 cells, about 107 cells to about 1010 cells, about 108 cells to about 109 cells, about 106 cells to about 108 cells, about 107 cells to about 109 cells, about 107 cells to about 1010 cells, about 107 cells to about 1011 cells, about 108 cells to about 1010 cells, about 108 cells to about 1011 cells, about 109 cells to about 1010 cells, about 109 cells to about 1011 cells, or about 1010 cells to about 1011 cells.
In some embodiments, the methods described herein involve administering a population of genetically engineered hematopoietic cells to a subject and administering one or more immunotherapeutic agents (e.g., cytotoxic agents). As will be appreciated by one of ordinary skill in the art, the immunotherapeutic agents can be of the same or different type (e.g., therapeutic antibodies, populations of immune cells expressing chimeric antigen receptor(s), and/or antibody-drug conjugates).
In some embodiments, the cytotoxic agent comprising an epitope binding fragment that binds an epitope of a cell-surface protein (e.g., immune cells expressing a chimeric receptor as described herein) is administered prior to administration of the hematopoietic cells. In some embodiments, the agent comprising an epitope binding fragment that binds an epitope of a cell-surface protein (e.g., immune cells expressing a chimeric receptor as described herein) is administered at least about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 3 months, 4 months, 5 months, 6 months or more prior to administration of the hematopoietic cells.
Alternatively, in some embodiments, the hematopoietic cells are administered prior to the cytotoxic agent comprising an epitope binding fragment that binds an epitope of the cell-surface protein (e.g., immune cells expressing a chimeric receptor as described herein). In some embodiments, the population of hematopoietic cells is administered at least about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 3 months, 4 months, 5 months, 6 months or more prior to administration of the cytotoxic agent comprising an epitope binding fragment that binds to an epitope of the cell-surface protein.
In some embodiments, the cytotoxic agent targeting the cell-surface protein and the population of hematopoietic cells are administered at substantially the same time. In some embodiments, the cytotoxic agent targeting the cell-surface protein is administered and the patient is assessed for a period of time, after which the population of hematopoietic cells is administered. In some embodiments, the population of hematopoietic cells is administered and the patient is assessed for a period of time, after which the cytotoxic agent targeting the cell-surface protein is administered.
Also within the scope of the present disclosure are multiple administrations (e.g., doses) of the cytotoxic agents and/or populations of hematopoietic cells. In some embodiments, the cytotoxic agents and/or populations of hematopoietic cells are administered to the subject once. In some embodiments, cytotoxic agents and/or populations of hematopoietic cells are administered to the subject more than once (e.g., at least 2, 3, 4, 5, or more times). In some embodiments, the cytotoxic agents and/or populations of hematopoietic cells are administered to the subject at a regular interval, e.g., every six months.
Examples of routes of administration include intravenous, infusion, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
Any of the methods described herein can be for the treatment of a hematological malignancy in a subject. The term “treat” or “treatment” or “treating” or “to treat” as used herein refers to therapeutic measures that aim to relieve, slow down progression of, lessen symptoms of, and/or halt progression of a pathologic condition or disorder. Thus, those in need of treatment include those already with the disorder. For example, in some instances, treating a cancer means stabilizing progression of the cancer. In some instances, treating a cancer means slowing down progression of the cancer. In some instances, treating a cancer means halting progression of the cancer. In some instances, treating a cancer means shrinking the cancer size. In some instances, treating a cancer means increasing the overall survival of the subject diagnosed with the cancer. Methods of assessing the progression of a cancer are known in the art and include, for example, evaluation of target lesions using imaging (e.g., X-ray, computerized tomography scan, magnetic resonance imaging, caliper measurement, or positron emission tomography scan), cytology or histology, or expression of tumor marker(s) (see, e.g., Eisenhauer et al., 2009, European Journal of Cancer 45:228-247 and Schwartz et al., 2016, European Journal of Cancer 62:132-137; each of which is incorporated by reference herein in its entirety).
In some embodiments, the subject is a human subject having a hematopoietic malignancy. As used herein a hematopoietic malignancy refers to a malignant abnormality involving hematopoietic cells (e.g., blood cells, including progenitor and stem cells). Examples of hematopoietic malignancies include, without limitation, Hodgkin's lymphoma, non-Hodgkin's lymphoma, leukemia, or multiple myeloma. Exemplary leukemias include, without limitation, acute myeloid leukemia, acute lymphoid leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia or chronic lymphoblastic leukemia, and chronic lymphoid leukemia.
In some embodiments, cells involved in the hematopoietic malignancy are resistant to conventional or standard therapeutics used to treat the malignancy. For example, the cells (e.g., cancer cells) can be resistant to a chemotherapeutic agent and/or CAR T cells used to treat the malignancy.
In some instances, the hematopoietic malignancies include: B-lymphoblastic leukemia (B-ALL), acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or Blastic Plasmacytoid Dendritic Cell Leukemia (BPCDN).
Any of the immune cells expressing chimeric receptors described herein can be administered in a pharmaceutically acceptable carrier or excipient as a pharmaceutical composition.
The phrase “pharmaceutically acceptable,” as used in connection with compositions and/or cells of the present disclosure, refers to molecular entities and other ingredients of such compositions that are physiologically tolerable and do not typically produce untoward reactions when administered to a mammal (e.g., a human). Preferably, as used herein, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in mammals, and more particularly in humans. “Acceptable” means that the carrier is compatible with the active ingredient of the composition (e.g., the nucleic acids, vectors, cells, or therapeutic antibodies) and does not negatively affect the subject to which the composition(s) are administered. Any of the pharmaceutical compositions and/or cells to be used in the present methods can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formations or aqueous solutions.
Pharmaceutically acceptable carriers, including buffers, are well known in the art, and can comprise phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; amino acids; hydrophobic polymers; monosaccharides; disaccharides; and other carbohydrates; metal complexes; and/or non-ionic surfactants. See, e.g. Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Williams and Wilkins, Ed. K. E. Hoover.
Also within the scope of the present disclosure are kits for use in treating hematopoietic malignancy. Such a kit can comprise the genetically engineered hematopoietic cells such as HSPCs, and optionally one or more cytotoxic agents targeting cell-surface antigens, the genes of which are edited in the hematopoietic cells. Such kits can include a container comprising a first pharmaceutical composition that comprises any of the genetically engineered hematopoietic cells as described herein, and optionally one or more additional containers comprising one or more cytotoxic agents (e.g., immune cells expressing chimeric receptors described herein) targeting the cell-surface antigens as also described herein.
In some embodiments, the kit can comprise instructions for use in any of the methods described herein. The included instructions can comprise a description of administration of the genetically engineered hematopoietic cells and optionally descriptions of administration of the one or more cytotoxic agents to a subject to achieve the intended activity in a subject. The kit can further comprise a description of selecting a subject suitable for treatment based on identifying whether the subject is in need of the treatment. In some embodiments, the instructions comprise a description of administering the genetically engineered hematopoietic cells and optionally the one or more cytotoxic agents to a subject who is in need of the treatment.
The instructions relating to the use of the genetically engineered hematopoietic cells and optionally the cytotoxic agents described herein generally include information as to dosage, dosing schedule, and route of administration for the intended treatment. The containers can be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. Instructions supplied in the kits of the disclosure are typically written instructions on a label or package insert. The label or package insert indicates that the pharmaceutical compositions are used for treating, delaying the onset, and/or alleviating a disease or disorder in a subject.
The kits provided herein are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging, and the like. Also contemplated are packages for use in combination with a specific device, such as an inhaler, nasal administration device, or an infusion device. A kit can have a sterile access port (for example, the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The container can also have a sterile access port. At least one active agent in the pharmaceutical composition is a chimeric receptor variants as described herein.
Kits optionally can provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container. In some embodiment, the disclosure provides articles of manufacture comprising contents of the kits described above.
The following examples are provided to better illustrate the claimed invention and are not to be interpreted as limiting the scope of the invention. To the extent that specific materials are mentioned, it is merely for purposes of illustration and is not intended to limit the invention. One skilled in the art can develop equivalent means or reactants without the exercise of inventive capacity and without departing from the scope of the invention.
WT-Cas9 and NG-Cas9 base editor plasmids were obtained through Addgene (plasmid #138495, 138491). SpRY-ABE8e-V106W 3xNLS and other base editor variants (see complete list below) were cloned using NEB HiFi assembly master mix and synthesized dsDNA inserts (IDT gBlocks). Single amino acid changes (i.e., K918N) or deletions (Blackjack variants) were introduced through standard site-specific mutagenesis techniques. When required, sgRNAs were cloned in a pLentiguide-Puro backbone (Addgene) or a pLKO-mTagBFP2 backbone (cloned) using the BsmBI restriction enzyme and annealed and phosphorylated DNA oligos with desired spacer sequence. Plasmid maxipreps were purified with Mackarey Nagel NucleoBond Xtra Maxi kit.
In order to evaluate the binding affinity of the mutated receptors for their ligand, a fluorescent ligand binding assay was developed. Human SCF and FLT3L (Peprotech) were conjugated with Alexa Fluor 488 Antibody Labeling Kit (Invitrogen cat. A20181) according to manufacturer's recommendations. Cells expressing either FLT3 or KIT variants were incubated at room temperature for 15 min with FcR-blocking reagent (Miltenyi 130-059-901), LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967), a control antibody (KIT 104D2 PE-Cy7, Biolegend, or FLT3 BV10A4 PE-Cy7, Biolegend) and the respective AF488-conjugated ligand. Samples were washed with PBS+2% FBS and analyzed with a BD Fortessa flow cytometer.
As K562 cells do not constitutively express FLT3 or CD123, these cells were engineered to over-express either FLT3, CD123, KIT or any combinations of the three genes from their endogenous genomic locus to serve as a model for gene editing approaches on these genes. In order to obtain constitutive strong expression of these genes, a full human EF1 alpha promoter was integrated upstream of the transcriptional start site of the FLT3, CD123 or KIT genes through CRISPR-Cas9 (FLT3, KIT) or CRISPR-AsCas12a (CD123) gene editing strategies (
K562 cells overexpressing FLT3/CD123 variants or NIH-3T3 or HEK-293T cells overexpressing KIT were cultured overnight in medium without FBS (serum starvation) and then 1 million cells from each condition were either stimulated or not with 100 ng/ml FLT3L, IL3 or SCF (depending on the evaluated receptor) for 5′ at 37° C. Cells were then washed 3× with ice cold PBS, lysed and proteins were extracted for western blot (Cell Extraction Buffer, ThermoFisher FNN0011+1 mM PMSF). After quantification with a BCA assay, protein extracts were mixed with Laemmli loading dye (Biorad 161-0747) and run on a Novex Tris-Glycine Gel (Invitrogen). After transfer, membranes were blocked with 5% w/v BSA in TBST and incubated with primary antibodies recognizing pKIT (Y719, Cell Signaling 3391T) or pFLT3 (Tyr589/591 clone 30D4, Cell Signaling 3464S) overnight at 4° C. After washing, membranes were incubated with Anti-rabbit IgG, HRP-linked Antibody (Cell Signaling 7074) for 1 hour at room temperature and later with SuperSignal West Femto chemiluminescent HRP-substrate (Thermo Scientific 34096) and analyzed with an ImageQuant LAS4000. The same membranes were subsequently incubated with Restore (ThermoScientific #21059) stripping buffer for 20 minutes at room temperature for a secondary staining with anti-KIT (clone 1C5, Invitrogen MA5-15894) or anti FLT3 (clone OTI7D6, Origene TA808157) primary antibodies with the same procedure described above. Secondary Ab staining was performed with anti-rabbit or anti-mouse IgG HRP-conjugated Abs according to the primary Ab and membranes were developed and acquired as above. Anti-actin staining was used as internal control for loading normalization.
Edited cell lines were evaluated by flow cytometry at 72 hours post editing and stained with antibodies clones either binding to the therapeutic epitope or an unrelated epitope to serve as control antibody for surface expression of the edited protein. For FLT3 editing, cells were incubated with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, FLT3 BV10A4 PE-Cy7 2/100 (Biolegend 313314) and FLT3 4G8 BV711 (BD 563908). For CD123 editing, cells were incubated with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, CD123 9F5 PE or BV421 (BD 555644) 1/100, CD123 7G3 BV711 or BV421 (BD 740722) 1/100. For KIT editing, cells were incubated with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, KIT 104D2 PE-Cy7 or BV711 2/100 (Biolegend 313212), KIT Fab79D AF488 or PE (Creative Biolabs) 2/100. Staining was performed at 4° C. for 30 minutes with 100 μL/sample volume. To evaluate the stem cell phenotype of cultured human CD34+ HSPCs, cells were harvested, resuspended in 100 μL PBS and stained with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, CD34 BV421 (Biolegend) 1.5/100, CD90 APC (BD) 3.5/100, CD45RA APC-Cy7 (Biolegend) 3.5/100, CD133/2 PE (Miltenyi) 4/100. Staining was performed at 4° C. for 30 minutes with 100 μL/sample volume. Samples were analyzed on a 4- or 5-laser BD Fortessa flow cytometer.
Peripheral blood from xeno-transplanted NSG mice was collected in 1.5 mL Eppendorf tubes with 10 μL 0.5M EDTA and stained with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, human CD45 BV786 (Biolegend) 1.5/100, mouse CD45 BV570 (Biolegend), CD34 BV421 (Biolegend) 1.5/100, CD19 BV650 (Biolegend) 3/100, CD3 BV711 (Biolegend) 2/100, CD33 BB515 (BD) 2/100 for 15 minutes at room temperature. Blood samples were then lysed with ACK reagent (StemCell technologies) for 5 minutes at room temperature and washed twice in PBS+2% FBS. Samples were analyzed on a 4-laser BD Fortessa flow cytometer.
Bone marrow from xeno-transplanted mice was obtained through crushing the hind limbs bones in a mortar, filtration through a 40-um cell strainer and resuspending the cells in Miltenyi MACS running buffer. A fraction of the cells was stained for flow cytometry analysis: with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 uL, 7-AAD (BD Pharmigen) 3/100, human CD45 BV786 (Biolegend) 1.5/100, mouse CD45 BV570 (Biolegend), CD34 BV421 (Biolegend) 1.5/100, CD19 BV650 (Biolegend) or CD19 BV605 (Biolegend) 3/100, CD3 BV711 (Biolegend) or CD3 PE-Cy7 (Biolegend) 2/100CD33 BB515 (BD), CD38 BV480 (BD) 1.5/100 or CD38 BUV396 (BD) 2/100, FLT3 BV10A4 PE-Cy7 (Biolegend), CD123 9F5 PE (BD), CD90 APC (BD) 3.5/100, CD45RA APC-Cy7 (Biolegend) 3/100 for 30 minutes at 4° C. Spleens were smashed on a 40 μm cell strainer and resuspended in Miltenyi MACS running buffer. Retrieved cells were stained with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 uL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, human CD45 Pacific Blue (Biolegend) 1.5/100, mouse CD45 BV570 (Biolegend), CD3 BV711 (Biolegend) 2/100, EGFR AF488 (R&D) 1.5/100, CD62L PE (BD) 2/100, CD4 APC (BD) 2/100, CD8 BV750 (Biolegend) 2/100, CD45RA APC-Cy7 (Biolegend) 3/100, CD69 PerCP-Cy5.5 (Biolegend) 3/100 for 30 minutes at 4° C. Samples were analyzed on a 4-laser BD Fortessa flow cytometer. In some experiments, BM cells were stained with hCD45 BV786, mCD45 PerCP-Cy5.5, CD3 PE-Cy5, CD7 AF700, CD10 BUV737, CD11c BUV661, CD14 BV510, CD19 BV605, CD33 PE-Cy7, CD38 BUV396, CD45RA APC-Cy7, CD56 BUV496, CD90 APC, FLT3 PE or BV711 and either KIT BV711 or CD123 PE antibodies with the addition of 50 uL/sample Brilliant stain buffer (BD cat.no. 659611).
gDNA Extraction, PCR Amplification and Sanger Sequencing
Genomic DNA was extracted from dry pellet samples using Qiagen DNeasy Blood & Tissue Kit or Lucigen QuickExtract reagent, quantified by Nanodrop 8000 and the sequences of interest were amplified by PCR using Promega GoTaq G2 and the respective primers. PCR products were purified using Promega SV Wizard Gel and PCR Clean-Up system and sent for Sanger sequencing through Genewiz. Base editing efficiencies were calculated from Sanger traces by deconvolution with the EditR R package using a custom script for high-throughput sample analysis.
Colony forming Unit assays (CFU) were performed by plating 1000 CD34+ cells/well, for in vitro CD34+ HSPCs experiments, or 25000 total bone marrow cells/wells, for xeno-transplanted BM-derived assays, unless stated otherwise. Cells were resuspended in Methocult H4034 media (StemCell cat.no. 04034) and plated in SmartDish meniscus-free 6-well plates. Wells were imaged after 2 weeks using StemCell STEMvision system. For flow cytometry analysis, methylcellulose media was softened with warm PBS, collected and washed twice before analysis.
In order to design mutated receptor variants which would not be recognized by selected monoclonal antibody clones, the epitopes targeted by different mAb were identified according to available information in the literature or by screening individual mutated receptor variants or mutation libraries. The overall goal was the identification of minimally modified target variants that preserve surface expression, gene regulation and signal transduction functionality while lacking recognition by selected therapeutic antibody clones (
FLT3 is a type III tyrosine kinase receptors composed by i) an extracellular ligand binding domain, characterized by the presence of five immunoglobulin-like domains; ii) a single spanning transmembrane region; iii) an intracellular part containing a split tyrosine kinase domain. The first 3 extracellular domains are involved in the binding with its dimeric ligand, FLT3-ligand (FLT3L), and this interaction induced the dimerization of the receptor. After dimerization, FLT3 activation is mediated by close positioning of the intracellular tyrosine kinase domains to each other, which facilitates their subsequent transphosphorylation.
Alignments show that mouse and human FLT3/FLT3L are 85.5% identical at the amino-acid level and that mouse and human FLT3 IgG-like domain 4 are 82% identical (95.5% similar) at the amino-acid level (
As a second step, the 16 mutations introduced to generate eFLT3-01 (SEQ ID NO: 49), were separated in two pools of mutations by their genomic localization within either FLT3 exon 9 or exon 10 (which together encode for FLT3 extracellular domain 4). Overexpression of these two FLT3 variants showed that the mutations restricted to FLT3 exon 9 (SEQ ID NO: 50) were sufficient to abrogate anti FLT3 clone 4G8, similarly to SEQ ID NO: 49. The FLT3 variant with mutations limited to exon 10, did not abrogate clone 4G8 binding.
To identify the key residues involved in the anti-FLT3 antibody FLT3 4G8 clone recognition of FLT3 extracellular domain 4, a combinatorial library was designed with all 16 previously identified residues either wild-type or mutated and cloned in a sleeping beauty transposon transfer vector under a EF1 alpha promoter (GeneScript-
Based on the available literature, anti-KIT antibody clone Fab79D recognizes putative amino-acid contact points in the extracellular domain 4 of KIT (
To expand these findings, a comprehensive library approach to further define alternative codons involved in Fab79D binding was designed. A degenerated library, where each codon within KIT extracellular domain 4 was composed of degenerated bases (NNN), was cloned in a sleeping beauty transfer plasmid expressing the human KIT cDNA, an mTagBFP reporter and puromycin resistance (
For CD123 (CD123), it has been reported in the literature that anti-CD123 antibody clones 7G3 and its humanized version, CSL362, recognize putative amino-acid contact points within CD123 N-terminal domain (
To engineer the target epitope within CD123 extracellular domain, a direct base editor screening approach was employed by designing a set of sgRNAs targeting 150, E51, Y58, S59, R84, P88 or P89 in the CD123 N-terminal domain (
A second round of sgRNA screening was carried out including additional sequences in close proximity to the sgRNAs identified in the previous round and were tested with several BE, including variants with relaxed PAM specificity (evo-APOBEC1-BE4, NG-EA-BE4max, NG-A3A-BE5, SpRY-evo-APOBEC1-BE4-SEQ ID NO: 8 and SEQ ID NO: 11, NG-ABE8e, SpRY-ABE8e-V106E-SEQ ID NO: 7 and SEQ ID NO: 10, LbCas12a-ABE8e-
To test if the FLT3 variants were selectively resistant to FLT3-targeting CAR-T cells, an in vitro killing assay was performed.
A III-generation lentiviral construct expressing a 2nd generation, FLT3-specific 4G8 clone chimeric antigen receptor with CD28 transmembrane region and CD28 co-stimulatory domain under a constitutive hPGK promoter was cloned using synthesized dsDNA fragments (IDT gBlocks). An antisense cassette expressing a truncated variant of the human EGFR cDNA under a minimal-CMV promoter was included to serve as marker of transduction and safety switch for in vivo depletion by using anti-EGFR antibody Cetuximab. VSV-G pseudotyped self-inactivating lentiviral particles were prepared according to published methods by calcium-phosphate transient co-transfection of 5 plasmids in HEK-293T cells (transfer vector, pMD2, pMDL-RRE, PREV and pAdvantage plasmids). Viral particles-containing supernatants were concentrated 500-fold by ultracentrifugation (20000 rpm at 20 C for 2 hours) and resuspended in PBS. Concentrated LVs were titrated by transducing 293T cells at different concentrations and calculating the transduction efficiency by flow cytometry or ddPCR.
Peripheral blood mononuclear cells (PBMC) were isolated by ficoll gradient separation from whole blood. After estimation of T cell fraction by flow cytometry, either freshly isolated or thawed PBMC were incubated with CD3-CD28 Dynabeads at 3:1 bead: T cell ratio (Gibco 11131D) for 45 min at room temperature on slow agitation and then magnetically separated (DynaMag-5 Magnet, Invitrogen 12303D). Positively selected cells were cultured with Dynabeads at 1 M/mL in IMDM supplemented with 10% FBS, 1% P/S, human IL-7 5 ng/mL (Peprotech) and human IL-15 5 ng/ml (Peprotech). 48h after the start of Dynabeads stimulation, T cells were transduced at MOI 5 to MOI 10 depending on the experiment with lentiviral particles encoding for the CAR of choice. Dynabeads were removed from culture by magnetic separation at day 7 since the start of the stimulation, and T cells were expanded for an additional 5-7 days in IMDM supplemented with 10% FBS, 1% P/S, human IL-7 5 ng/ml (Peprotech) and human IL-15 5 ng/ml (Peprotech). T cell phenotype and transduction efficiency (by EGFR surface staining) was evaluated periodically by flow cytometry. Expanded CAR-T cells or untransduced T cells were either used for killing assays, in vivo administration or vitally frozen after 12-14 days since the start of the stimulation.
K562 cells (either unmodified, base edited or overexpressing a receptor variant after sleeping-beauty transduction) were plated in a 96-well plate (25000 target cells/well). Anti-FLT3 CAR-T cells were generated by transducing peripheral blood mononuclear cells (PBMC with a lentiviral vector encoding for a 2nd generation 4G8-CAR construct with CD28 costimulatory domain, and co-expressing an EGFRt (truncated EGFR) safety switch (see protocol above). FLT3-targeting 4G8 CAR-T cells or untransduced T cells were marked with CellTrace yellow (Invitrogen C34567) according to the manufacturer's recommendations. FLT3-targeting 4G8 CAR-T cells or untransduced T cells were then co-plated at different effector: target ratios (E:T ratio), typically 10, 5, 2.5, 1.25, 0.625 in the same wells and incubated at 37° C. with 5% CO2 in a humidified incubator. After 4 hours, 50% of the culture volume was harvested for flow cytometry analysis by staining with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, FLT3 BV10A4 PE-Cy7 2/100 (Biolegend 313314), CD4 APC (BD) 2/100, CD8 BV750 (Biolegend) 2/100, CD107a BV711 (Biolegend) 2/100, CD69 PerCP-Cy5.5 (Biolegend) 2/100. The staining mix included flow counting beads to normalize cell counts (Biolegend Precision Count Beads). Cells were than washed and resuspended in AnnexinV binding buffer (Biolegend) supplemented with AnnexinV FITC (Biolegend) 3/100. Samples were analyzed on a 4- or 5-laser BD Fortessa flow cytometer. The remaining culture volume was evaluated at 48 hours post plating by flow cytometry stained with FcR-blocking reagent (Miltenyi 130-059-901) 2/100 μL, LIVE/DEAD Fixable Yellow Dead Cell Stain (Invitrogen L34967) 1/1000, FLT3 BV10A4 PE-Cy7 2/100 (Biolegend 313314), CD4 APC (BD) 2/100, CD8 BV750 (Biolegend) 2/100, CCR7 BV421 (Biolegend) 2/100, CD45RA APC-Cy7 (Biolegend) 3/100, CD33 PerCP-Cy5.5 (Biolegend). Cells were than washed and resuspended in AnnexinV binding buffer (Biolegend) supplemented with AnnexinV FITC (Biolegend) 3/100.
To test if FLT3 N399D can be introduced by homology directed repair, a CRISPR Cas homology directed repair (HDR) strategy was designed. SpCas9 or AsCas12a nucleases in combination with several gRNAs targeting the FLT3 exon 9 locus were tested in combination with and 200-nt long single strand oligo-deoxynucleotide (ssODN) as donors template for HDR. Each donor templates included selected silent mutations in bystander amino-acids to reduce the risk of re-cutting by the CRISPR-Cas9 RNP nuclease complex after successful DNA repair. The reverse complementary of each ssODN donors (termed A, C, H, F) were also tested. The sequences of the ssODN template donors are reported as SEQ ID NOS: 40 through 43 (see above in Table 2). K562 reporter cells overexpressing FLT3 by targeted integration of an EF1-a promoter upstream of the endogenous FLT3 locus were electroporated using Lonza 4D-Nucleofector system in SF solution supplemented with 50 pmol of 3xNLS Cas9 nuclease (IDT) complexed with 62.5 pmol annealed trRNA:gRNA or 50 pmol AsCas12a Ultra (IDT) complexed with 62.5 pmol sgRNA according to experimental condition. Cas9 gRNA e9-4-NGG was tested in combination with ssODN-A and C and their reverse complements (5 μM final concentration). Cas12a gRNAs e9-15-TTTV and e9-16-TTTV were tested in combination with ssODN-H and F and their reverse complements (5 uM final concentration). IDT HDR-enhancer 0.2 uL/20 uL was included in the electroporation reaction (30 uM). The outcome of the editing procedure was evaluated by flow cytometry 72 h after electroporation.
In order to introduce the desired single codon change in the FLT3 locus with high efficiency and low toxicity, without the introduction of double-strand DNA breaks, we tested CRISPR-Cas base editing. We designed a panel of sgRNAs that were predicted to introduce the N399D or N399G mutation in combination with adenine base editors. CRISPR-Cas9 base editor ABE8e (TadA-8e V106W) was selected for the development of the editing strategy and further optimized by mutating the Cas9 nickase protein to relax the PAM specificity in order to allow editing in the absence of conventional NGG PAM. To this end, both NG-SpCas9 and SpRY-Cas9 variants of the base editor were cloned. To further increase the efficiency, a 3rd nuclear localization site (NLS) was fused to the C-terminal portion of the protein. Unless stated otherwise, base editing experiments were performed by electroporation of reporter K562 overexpressing the FLT3 gene with 500 ng of base editor plasmid and either 300-360 pmol of sgRNA (Integrated DNA Technologies). Cells were then cultured and samples for genomic DNA and flow cytometry analysis were harvested 72 h after editing.
To improve efficiency of base-editing of the N399 codon, tailored editing window positioning was achieved by screening additional sgRNAs in combination with near PAM-less SpRY-Cas9 variant.
Sanger sequencing of base edited cells from Example 5 revealed A to G editing of both adenines within the N399 codon (sequence: AAC) with potential generation of either N399D or N399G mutations. To further test if FLT3 variants containing a mutation at N399 position still preserve physiological FLT3L binding, fluorescent ligand binding assays were performed.
In order to translate the base editing procedure to primary cells, a suitable delivery method for base editors needs to be developed, as bacterial plasmid transfection is reported to be toxic for stem cells.
We employed base editor mRNA produced by in vitro transcription to translate the base editing protocol to human CD34+ HSPC. Functional mRNA encoding for adenine base editors (SpRY-ABE8e-V106W 3xNLS or SpRY-K918N-ABE8e-V106W 3xNLS) were produced by in vitro transcription (IVT) using MEGAscript T7 Transcription Kit (Invitrogen AM1333) or NEB T7 HiScribe kit (E2040S) and a custom plasmid (SEQ ID NO: 81; see
To confirm that FLT3 base editing of primary human CD34+ hematopoietic stem and progenitor cells is feasible, an in vitro base editing and expansion culture experiment was performed. Mobilized peripheral blood-derived CD34+ HSPCs were thawed and cultured at 0.5-0.75 million/mL in StemCell SFEMII medium supplemented with 1% Penicillin/Streptomycin, SCF 100 ng/ml (Peprotech), FTL3L 100 ng/ml (Peprotech), TPO 50 ng/mL (Peprotech), Stemregenin-1 0.75 uM (StemCell technologies), UM171 35 nM (Selleckhem). 0.15-0.25 million (M) HSPCs were electroporated either 48 hours after thawing using Lonza 4D-Nucleofector system in P3 electroporation solution (Lonza) supplemented with 2.5-7.5 ug base editor mRNA (SpRY-ABE8e-V106W) and sgRNA (Integrated DNA Technologies) 250-450 pmol per 20 uL reaction. Cells were cultured in the aforementioned medium for 5-7 days. To test for specific resistance to CAR-T cell killing, edited or unedited CD34+ cells were expanded in vitro for 3 days post editing and then co-cultured with either anti-FLT3 CAR-T cells or untransduced T cells.
Editing efficiencies by Sanger sequencing at day6 post electroporation are reported in
To test if base edited cells obtained from the experiments done in
Similarly, to test if base edited human hematopoietic stem and progenitor cells are resistant to FLT3-targeting immunotherapies, CD34+ edited in the experiment performed in
To further confirm the protective role against on-target killing mediated by anti-FLT3 CAR-T cells, an in vivo xenotransplantation experiment in immunodeficient NSG mice was performed (
Hematopoietic stem and progenitor cell frequency (CD34+CD38−) and absolute abundance in the bone marrow at sacrifice are reported in
To further evaluate the protection of CD34+ HSPCs by FLT3 base editing of N399 against CAR-T cell killing, a second experiment in vivo experiment was performed in NBSGW mice, which allow xeno-transplantation and engraftment of human HSPCs without irradiation and, thanks to a hypomorphic Kit mutation in murine HSPCs, (homozygous W41 allele) higher levels of human hematopoietic reconstitution.
Similar to FLT3-base editing experiments, we introduced the S59P mutation using SpRY-ABE8e-V106W mRNA and CD123 gRNA-N in human CD34+ HSPCs, either alone or in combination with FLT3 N399 base editing. Mobilized peripheral blood-derived CD34+ HSPCs were thawed and cultured at 0.5-0.75 million/mL in StemCell SFEMII medium supplemented with 1% Penicillin/Streptomycin, SCF 100 ng/ml (Peprotech), FTL3L 100 ng/ml (Peprotech), TPO 50 ng/ml (Peprotech), Stemregenin-1 0.75 uM (StemCell technologies), UM171 35 nM (Selleckhem). 0.15-0.25 million (M) HSPCs were electroporated either 48 hours after thawing using Lonza 4D-Nucleofector system in P3 electroporation solution (Lonza) supplemented with 5-7.5 ug base editor mRNA (SpRY-ABE8e-V106W) and sgRNA (Integrated DNA Technologies) 420-450 pmol per 20 uL reaction. Cells were cultured in the aforementioned medium for 7 days.
To see if editing efficiency of CD123 S59 codon could be improved, a Cas9 protein containing the K918N mutation, which has been associated with improved Cas9 catalytic activity was tested in combination with our SpRY-ABE8e design. As shown in
In order to further improve CD123 editing efficiency, two additional sgRNAs (CD123-gRNA-R and its 21-bp long version, CD123-gRNA-R21) were cloned alongside the benchmark CD123-gRNA-N and FLT3-gRNA-18 in a pHKO-mTagBFP2 plasmid under a human U6 promoter.
Improvement in CD34+ HSPCs base editing efficiencies for FLT3 and CD123 loci using SpRY-K918N-ABE8e-V106W in vitro transcribed mRNA was achieved through optimization of multiple parameters (mRNA preparation, K918N mutation introduction, sgRNA selection). An exemplary experiment is reported in
Using the same experimental setting as Example 12, we tested whether H378R mutation could be introduced through base editing in human CD34+ HSPCs. By electroporating SpRY-K918N-ABE8e-V106W in vitro transcribed mRNA (4 ug/20 μL electroporation volume) and KIT-gRNA-Y (SEQ ID NO: 37), editing efficiency up to 60% was achieved.
Rationally designed anti-FLT3 and CD123 bispecific chimeric antigen receptors should result in potent antileukemia efficacy.
As targets for our epitope engineering strategy, we selected the cytokine receptors FLT3, KIT and CD123 (IL3RA). Fms-like tyrosine kinase 3 (FLT3, CD135) and proto-oncogene c-KIT (KIT, CD117) are class III receptor tyrosine kinase which are expressed, either in wild type (WT) or mutated form, in 93% and 85% of AML cases, respectively32-37. CD123 is the alpha subunit of the IL-3 receptor (IL3RA), a type I cytokine receptor found on the surface of >75% of AML cases and overexpressed on the surface of leukemic stem cells38-40. These genes are present at various stages of normal hematopoietic development and their overexpression on AML cells is associated with poor prognosis, with higher incidence of relapse after HSCT and a lower overall survival rate both in adult and pediatric patients33,36,41-43. To develop our approach, we selected monoclonal antibodies (mAb) currently under evaluation for the development of anti-AML immunotherapies: clone 4G822,44 (FLT3), Fab-79D21,45 (KIT) and 7G346-48 (CD123). Previous studies reported that 4G8 recognizes FLT3 extracellular domain (ECD) 4, while clone BV10A4 recognizes an unrelated epitope within ECD2 and therefore can serve as control to assess FLT3 surface expression44. Since 4G8 was generated by immunizing BALB/c mice with human FLT3 transfected cell lines, we reasoned that 4G8 recognizes a human-specific epitope, despite the high degree of homology (85.8% identity and 91.5% similarity) and FLT3 ligand (FLT3L) cross-reactivity between human and mouse FLT349.
We first confirmed that a chimeric human FLT3 with substitution of ECD4 with its murine ortholog (16 codon changes) results in loss of 4G8 binding without affecting FLT3L binding and intracellular kinase phosphorylation (
Next, we evaluated gene editing strategies to introduce the N399D substitution. To easily evaluate the outcomes of our genome engineering procedures on cells that do not depend on FLT3 signaling, we generated K562 reporter cells that express FLT3 from their endogenous locus by targeted integration of an EF1α constitutive promoter upstream to the transcriptional start site (
Since epitope engineering can be achieved by the introduction of single point mutations, we reasoned that base editing (BE) could be a suitable and safer option for epitope editing by avoiding the need for DSB. The Asparagine in position 399 is encoded by an AAC codon, which can be converted to GAC (Aspartate) or GGC (Glycine) by Adenine Base Editing (ABE). We tested this hypothesis by electroporating the FLT3-reporter cells with several sgRNAs (in a 1-bp staggered fashion, with the target Adenine in position 3 to 9 of the protospacer) in combination with the advanced generation TadA-8e deaminase, linked either with SpCas9 nickase (NGG PAM) or Cas9 variants with relaxed PAM-specificity (NG-SpCas9n and SpRY-Cas9n,
A similar strategy was applied for the epitope mapping of Fab-79D, a KIT-targeting mAb which is reported to bind to KIT ECD4 and to block ligand-induced dimerization45. We first confirmed loss of Fab-79D binding by introducing 10 amino-acid changes (from KIT orthologs) at positions previously predicted as potential contact points with KIT ECD4 (F316S, M318V, I319K, V323I, 1334V, E360K, P363V, E366D, E376Q, H378R) and verified the preservation of stem cell factor (SCF) binding and intracellular kinase phosphorylation (
For CD123, both the epitope and the amino acid substitutions affecting the binding of therapeutic clone 7G3 (or its humanized counterpart, CSL362—Talacotuzumab) have been previously reported48. To develop our epitope-editing strategy, we designed a targeted BE screening on K562 reporter cells (
To precisely quantify the loss of affinity of our selected Abs for the epitope-engineered receptor, we transduced K562 cells by Sleeping Beauty transposase with each receptor variant (FLT3 WT, N399D, N399G; KIT WT, H378R; CD123 WT, S59P and Y58H-S59P) and observed almost complete absence of recognition of all tested variants also at saturating concentration of mAb for the WT receptor (>5000 ng/ml,
Overall, we concluded that epitope engineering of FLT3, KIT and CD123 is feasible and can be achieved with high efficiencies without the need for DSB by selecting appropriate combinations of gRNA and base editing enzymes.
As the selected targets are fundamental cytokine/growth factor receptors expressed on human HSPCs and have relevant roles in stem cell maintenance and lineage differentiation, we stringently assessed if our engineering procedure would alter receptor functionality. By using fluorescently conjugated FLT3L, SCF and IL-3 ligands, we confirmed comparable binding to their respective WT or epitope engineered receptors across all tested concentrations (1 to >1000 ng/ml;
Recent studies have shown that CAR-T cells generated from the anti-FLT3 clone 4G822, the anti-KIT Fab-79D21 or the anti-CD123 CSL362—the humanized variant of clone 7G348,51—mAbs have remarkable efficacy against human AML cells. To assess resistance of epitope engineered cells to targeted CAR-T therapy, we cloned the 4G8, Fab79D and CSL362 single-chain variable fragments (scFv) in a 2nd generation CAR construct with a CD28 costimulatory domain and used a lentiviral vector with a bidirectional promoter to co-express an optimized52 truncated EGFR selection/depletion marker (tEGFR;
By performing an in vitro killing assay on K562 reporter cells we found that, while the majority of cells overexpressing WT FLT3, KIT or CD123 were killed by their respective specific CAR-T (at E:T=10, <2% surviving cells compared to E:T=0), K562 cells expressing epitope-engineered variants were resistant to CAR-T cell killing (both as absolute counts and cell viability) across different effector: target ratios (E:T 0.625 to 10) and survived up to experiment termination (
Overall, these data provide a stringent validation that cells over-expressing the epitope engineered FLT3, KIT and CD123 variants are resistant to CAR-T cell recognition and killing.
To effectively introduce our nucleotide variants into the endogenous genes of human primary HSPCs, we optimized a base editing protocol on mobilized-peripheral blood (mPB)-derived CD34+ cells based on co-electroporation of a chemically modified sgRNA and in vitro transcribed (IVT) SpRY-ABE8e mRNA (
To confirm the resistance of FLT3N399 and CD123S59 epitope-engineered HSPCs to immunotherapies, we performed a killing assay by plating, 3-5 days after electroporation, edited HSPCs with 4G8 and CSL362 CAR-T cells at different E:T ratios. Specific killing by FLT3- and CD123 CAR-T was most pronounced within the CD45RA+ and the CD90+ subsets, respectively, which were thus used to evaluate the outcome of these experiments by absolute counts. While cells edited in a control site (AAVS1 safe genomic harbor) were eliminated by CAR-T cell co-culture, epitope-edited cells showed higher viability and absolute counts, similar to what we observed with K562 co-cultures (
To confirm preservation of functionality of the receptor engineered CD34+ HSPCs, treated cells were cultured for 4 additional days with increasing concentrations of their respective ligand and without other cytokines. At the end of culture, we observed dose-dependent HSPCs proliferative expansion for all 3 targets, with no difference between receptor-edited and AAVS1-edited controls except for CD123S59 BE at IL-3 concentrations of 1-10 ng/ml (
Overall, these data showed that epitope engineered HSPCs can be efficiently achieved by adenine base editor without affecting stem cell functionality and differentiation capacity.
In order to assess if FLT3 CAR-T cells can be effectively used to treat AML while sparing FLT3N399 hematopoiesis, we sequentially engrafted NBSGW mice with CD34+ HSPCs (either FLT3BE or AAVS1BE) and a human patient-derived AML xenograft (PDX-1), characterized by MLL-AF9 and FLT3-ITD mutations and previously transduced with a reporter gene—mNeonGreen− to facilitate its detection within the mixed hematopoiesis (
Overall, these data confirmed that in the NBSGW model, FLT3 CAR-T cell immunotherapy preferentially depletes B cell and progenitor subsets (GMP, LMPP) while FLT3N399 epitope editing confers protection to these subpopulations.
As done for FLT3 epitope editing, we performed xeno-transplantation of CD123S59 HSPCs into NBSGW mice and confirmed engraftment and multilineage repopulating capacity similar to AAVS1BE HSPCs (
Overall, these data showed a reduction of on-target toxicity induced by CD123 CAR-T cells on CD123S59 epitope edited hematopoiesis, which would otherwise result in depletion of the myeloid lineage, DCs and overall reduction in absolute counts of hematopoietic cells.
We reasoned that editing of two or more epitopes might enable even more effective immunotherapies by simultaneously attacking multiple AML targets without increasing hematopoietic toxicity. To assess the feasibility of multiple epitope-editing on primary HSPCs, we co-electroporated SpRY-ABE8e mRNA with two gRNAs on CD34+ cells and observed editing efficiencies comparable to those measured on the same targets after single BE, without overt increase in cellular toxicity (
To model the increased efficacy conferred by dual-targeting immunotherapies, we identified an AML PDX that is not effectively eradicated by 4G8 CAR-T cell treatment (PDX-2 characterized by MLL-AF10 and mutated TP53,
Overall, these data provide the proof-of-concept that multiplex epitope engineering can be efficiently obtained in HSPCs and enables potent multi-target immunotherapies with reduction of overlapping on-target off-tumor hematopoietic toxicities.
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
This application is the § 371 U.S. National Stage of International Application No. PCT/US2023/062757, filed Feb. 16, 2023, which claims the priority benefit of U.S. Provisional Application No. 63/311,707, filed Feb. 18, 2022, and to U.S. Provisional Application No. 63/426,138, filed Nov. 17, 2022, the contents of which are incorporated by reference in their entireties herein.
| Filing Document | Filing Date | Country | Kind |
|---|---|---|---|
| PCT/US2023/062757 | 2/16/2023 | WO |
| Number | Date | Country | |
|---|---|---|---|
| 63311707 | Feb 2022 | US | |
| 63426138 | Nov 2022 | US |