Introduction
Allogeneic hematopoietic stem cell (HSC) transplantation can be used to treat blood cancers, but it often leads to graft vs. host disease [
1]. Autologous HST transplantation would be better, and mobilization of HSCs can now be done much more effectively than in the past [
2]. However, the harvested patient cells could be contaminated to some extent with their tumor cells. A targeted method of purging HSCs could allow for safer autologous HSC transplantation [
3].
If the patient’s blood cancer cells in circulation, as well as the lymph nodes and bone marrow, are sequenced [
4], clonal mutations can be identified - i.e., mutations that are contained in all of their cancer cells. These mutations can be detected with molecular switches, and the cells can be eliminated using the effector module of said switches [
5,
6,
7].
This process can be done ex vivo, using transfection or transduction. It can also be done in vivo via mobilization of patient HSCs and dual adenoviral vector transduction [
8]. In this in vivo system, one adenoviral genome expresses Flpe recombinase and a hyperactive transposase to circularize and randomly insert the transgene cassette in the second into the HSC genome.
Purging Strategy
First, cancer cells in the patient’s bloodstream, lymph nodes, and bone marrow would be sequenced to determine his or her clonal mutations
4. After isolating mobilized CD
34+ cells from a patient’s bloodstream, an ex vivo vector could target CD34 for cell entry
2. Here, the early enrichment strategy for CD
34+ HSCs containing the gene vector, either as an integrated element or S/MAR-containing plasmid [
9], would rely on epitope base editing of the CD45 gene [
10]. Epitope base editing would shield HSCs containing the therapeutic DNA from an immunotoxin and be important for cancer therapy purposes [
11,
12,
13].
After purging and CD45 base editing, a replicating plasmid can be destroyed via CRISPR/Cas9 targeting of multiple plasmid loci. In the case of an integrated vector, a recombinase could excise the therapeutic DNA.
For in vivo purposes, mobilized HSCs would be targeted by the dual adenoviral vectors that can bind CD34
8. Here,
CD45 base editing would also be effected. Display of self-peptide based on CD47 on the adenoviral capsids [
14,
15] may preclude or reduce the need for innate immune suppression. Protein engineering approaches could shelter transduced HSCs from the adaptive immune system [
16,
17]. Alternatively, MHC class I and natural killer cell inhibitor proteins could be expressed by the vector during this process [
18]. Moreover, zinc finger base editors may be less immunogenic than TALE or CRISPR base editors [
19,
20]. The CRISPRa and base editor modules at least could be regulated by a recently developed, tetracycline-regulated RNA switch, as RNA elements would be non-immunogenic [
21].
A recombinase could also be utilized here for excision of the integrated transgene cassette after purging and CD45 base editing.
The ex vivo vector or adenoviral genome-derived transgene cassette would encode switches that can recognize the patient’s clonally mutated mRNA sequences and respond via activation of a toxin module that eliminates the cell [
5,
6,
7]. One example of a type of switch that could accomplish this is the reprogrammable ADAR sensor “RADAR”, a system developed recently that can even detect point mutations in mRNA [
22,
23]. It would also be programmed to express CRISPRa modules that upregulate the expression of the patient’s clonally mutated genes.
As described previously, CRISPRa expression could be halted via detection of the clonally mutated transcript somewhere other than the mutation site, as this would prevent long-term activation of the gene in non-cancerous cells [
7]. This site could be recognized by an RNA-binding “proximity” switch based on Pumby modules [
6,
24].
Blood Cancer Elimination Strategy
Base editing of the CD45 gene in the patient’s HSCs would allow anti-CD45 chimeric antigen receptor (CAR) T-cells to be intravenously administered and eliminate the remainder of the patient’s blood cancer cells [
25]. The CAR T-cells can be purged as well if necessary, and should have a base edited CD45 gene themselves to avoid fratricide. For acute myeloid leukemia, instead of
CD45 epitope editing, a combination of
FLT3,
CD123, and
KIT epitope editing can be exploited [
26]. The CAR T-cells could potentially be off-the-shelf, precluding tumor cell contamination issues [
18,
27]. They could be eliminated after treatment and re-administered later if necessary [
28]. An immunotoxin targeting CD45 could be used instead of CAR T-cells [
29]. It was demonstrated that the base edited CD45 gene product for the immunotoxin cross-referenced here [
10] was more biophysically optimal than the one generated earlier for CAR T-cell targeting [
25]. However, repeated injections may be required for immunotoxins, as opposed to autologous CAR T-cells, which can persist in circulation for long periods of time.
Conclusion
The purging strategy described in this piece could ensure that anti-CD45 immunotherapy involving base editing of patient HSCs can be employed to treat blood cancer without generating CD34+ cancer cells resistant to the therapy.
In the near future, multiplexed epitope editing of universal leukocyte markers other than CD45 could be performed to avoid the survival of escape variants.
This clonal mutation targeting strategy can be used in the context of solid tumors as well, using oncolytic vectors that require detection of said mutations for replication and hyper-virulence5,6,7. It may even be applicable to hematological malignancies like multiple myeloma, which forms solid tumors. Whether it can help treat others depends in part on whether it can efficiently reach cells in the bone marrow after intravenous injection.
Conflicts of Interest
None.
References
- Malard, F.; Holler, E.; Sandmaier, B.M.; et al. Acute graft-versus-host disease. Nat Rev Dis Primers 2023, 9, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Crees, Z.D.; Rettig, M.P.; Jayasinghe, R.G.; et al. Motixafortide and G-CSF to mobilize hematopoietic stem cells for autologous transplantation in multiple myeloma: a randomized phase 3 trial. Nat Med 2023, 29, 869–879. [Google Scholar] [CrossRef] [PubMed]
- Ishitsuka, K.; Nishikii, H.; Kimura, T.; et al. Purging myeloma cell contaminants and simultaneous expansion of peripheral blood-mobilized stem cells. Experimental Hematology 2024, 131, 104138. [Google Scholar] [CrossRef]
- Landau, D.A.; Carter, S.L.; Getz, G.; et al. Clonal evolution in hematologic malignancies and therapeutic implications. Leukemia 2014, 28, 34–43. [Google Scholar] [CrossRef]
- Renteln, M. Conditional replication of oncolytic viruses based on detection of oncogenic mRNA. Gene Ther 2018, 25, 1–3. [Google Scholar] [CrossRef]
- Renteln, M.A. Promoting Oncolytic Vector Replication with Switches that Detect Ubiquitous Mutations. CCTR 2024, 20, 40–52. [Google Scholar] [CrossRef]
- Renteln, M. Targeting Clonal Mutations with Synthetic Microbes. 2024. [CrossRef]
- Wang, H.; Georgakopoulou, A.; Zhang, W.; et al. HDAd6/35++ - A new helper-dependent adenovirus vector platform for in vivo transduction of hematopoietic stem cells. Mol Ther Methods Clin Dev 2023, 29, 213–226. [Google Scholar] [CrossRef]
- Bozza, M.; Green, E.W.; Espinet, E.; et al. Novel Non-integrating DNA Nano-S/MAR Vectors Restore Gene Function in Isogenic Patient-Derived Pancreatic Tumor Models. Molecular Therapy - Methods & Clinical Development 2020, 17, 957–968. [Google Scholar] [CrossRef]
- Garaudé, S.; Marone, R.; Lepore, R.; et al. Selective haematological cancer eradication with preserved haematopoiesis. Nature 2024, 630, 728–735. [Google Scholar] [CrossRef]
- Dever, D.P.; Bak, R.O.; Reinisch, A.; et al. CRISPR/Cas9 Beta-globin Gene Targeting in Human Hematopoietic Stem Cells. Nature 2016, 539, 384–389. [Google Scholar] [CrossRef]
- Haltalli, M.L.R.; Wilkinson, A.C.; Rodriguez-Fraticelli, A.; et al. Hematopoietic stem cell gene editing and expansion: State-of-the-art technologies and recent applications. Experimental Hematology 2022, 107, 9–13. [Google Scholar] [CrossRef] [PubMed]
- Meaker, G.A.; Wilkinson, A.C. Ex vivo hematopoietic stem cell expansion technologies: recent progress, applications, and open questions. Experimental Hematology 2024, 130. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, P.L.; Harada, T.; Christian, D.A.; et al. Minimal “Self” peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science 2013, 339, 971–975. [Google Scholar] [CrossRef] [PubMed]
- Milani, M.; Annoni, A.; Moalli, F.; et al. Phagocytosis-shielded lentiviral vectors improve liver gene therapy in nonhuman primates. Science Translational Medicine 2019, 11, eaav7325. [Google Scholar] [CrossRef] [PubMed]
- Ferdosi, S.R.; Ewaisha, R.; Moghadam, F.; et al. Multifunctional CRISPR-Cas9 with engineered immunosilenced human T cell epitopes. Nat Commun 2019, 10, 1842. [Google Scholar] [CrossRef]
- Hoyng, S.A.; Gnavi, S.; de Winter, F.; et al. Developing a potentially immunologically inert tetracycline-regulatable viral vector for gene therapy in the peripheral nerve. Gene Ther 2014, 21, 549–557. [Google Scholar] [CrossRef]
- Wang, X.; Cabrera, F.G.; Sharp, K.L.; et al. Engineering Tolerance toward Allogeneic CAR-T Cells by Regulation of MHC Surface Expression with Human Herpes Virus-8 Proteins. Molecular Therapy 2021, 29, 718–733. [Google Scholar] [CrossRef]
- Willis, J.C.W.; Silva-Pinheiro, P.; Widdup, L.; et al. Compact zinc finger base editors that edit mitochondrial or nuclear DNA in vitro and in vivo. Nat Commun 2022, 13, 7204. [Google Scholar] [CrossRef]
- Fauser, F.; Kadam, B.N.; Arangundy-Franklin, S.; et al. Compact zinc finger architecture utilizing toxin-derived cytidine deaminases for highly efficient base editing in human cells. Nat Commun 2024, 15, 1181. [Google Scholar] [CrossRef]
- Luo, L.; Jea, J.D.-Y.; Wang, Y.; et al. Control of mammalian gene expression by modulation of polyA signal cleavage at 5′ UTR. Nat Biotechnol 2024, 1–13. [Google Scholar] [CrossRef]
- Kaseniit, K.E.; Katz, N.; Kolber, N.S.; et al. Modular, programmable RNA sensing using ADAR editing in living cells. Nat Biotechnol 2023, 41, 482–487. [Google Scholar] [CrossRef] [PubMed]
- Gayet, R.V.; Ilia, K.; Razavi, S.; et al. Autocatalytic base editing for RNA-responsive translational control. Nat Commun 2023, 14, 1339. [Google Scholar] [CrossRef] [PubMed]
- Adamala, K.P.; Martin-Alarcon, D.A.; Boyden, E.S. Programmable RNA-binding protein composed of repeats of a single modular unit. Proc Natl Acad Sci USA 2016, 113, E2579–E2588. [Google Scholar] [CrossRef] [PubMed]
- Wellhausen, N.; O’Connell, R.P.; Lesch, S.; et al. Epitope base editing CD45 in hematopoietic cells enables universal blood cancer immune therapy. Sci Transl Med 2023, 15, eadi1145. [Google Scholar] [CrossRef]
- Casirati, G.; Cosentino, A.; Mucci, A.; et al. Epitope editing enables targeted immunotherapy of acute myeloid leukaemia. Nature 2023, 621, 404–414. [Google Scholar] [CrossRef]
- Jo, S.; Das, S.; Williams, A.; et al. Endowing universal CAR T-cell with immune-evasive properties using TALEN-gene editing. Nat Commun 2022, 13, 3453. [Google Scholar] [CrossRef]
- Stavrou, M.; Philip, B.; Traynor-White, C.; et al. A Rapamycin-Activated Caspase 9-Based Suicide Gene. Mol Ther 2018, 26, 1266–1276. [Google Scholar] [CrossRef]
- Hayal, T.B.; Wu, C.; Abraham, D.; et al. The Impact of CD45-Antibody-Drug Conjugate Conditioning on Clonal Dynamics and Immune Tolerance Post HSPC Transplantation in Rhesus Macaques. Blood 2023, 142, 3419. [Google Scholar] [CrossRef]
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).