<<

Review Article Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Induced pluripotent stem cells (iPSCs) based approaches for hematopoietic cancer therapy

Bardia Khandany BSc1, Mohammad Mehdi Heidari PhD1,*, Mehri Khatami PhD1

1. Department of Biology, Faculty of Science, Yazd University, Yazd, Iran. *Corresponding author: Dr Mohammad Mehdi Heidari, PhD, Department of Biology, Faculty of Science, Yazd University, Pajoohesh Street, Yazd, Iran. Email: [email protected].

Received: 23 Dcember 2018 Accepted: 02 March 2019

Abstract Induced pluripotent stem cells (iPSCs) are reprogrammed from somatic cells through numerous factors. Human induced pluripotent approaches are developing as a hopeful strategy to improve our knowledge of genetic association studies and the underlying molecular mechanisms. Rapid progression in stem cell therapy and cell provides compelling reasons for its feasibility for treating a wide range of diseases through the replacement of autologous cells. Continuous failure in embryonic stem cells (ESC) production and the dependency of iPSC on ectopic genes may be due to the inability to maintain the stability of the endogenous gene systems which are essential for creation of pluripotency state. With recent developments in the genome processing and human tissue culturing approaches as well as xenotransplantation, bioengineering, and genome editing, induced pluripotent stem cells offer the new opportunities for the study of human cancers. Most hematopoietic malignancies are originated from cells that are functionally heterogeneous and few of them are responsible for maintaining tumor state. The naming of these cancer stem cells are due to the quality characteristics of normal tissue stem cells, such as self-renewal, long term survival, and the ability to produce cells with more differentiated properties. The aim of present study was to focus on the recent progresses in the application of stem cell-based hematopoietic cancer, and to assess the benefits of treatment, opportunities, and shortcomings that can potentially help improve future efforts in experimental and clinical studies. Keywords: Gene therapy, Hematopoietic cancer, Induced pluripotent stem cells

Introduction conditions that allowed specialized adult The human body comprises of over two cells to be genetically “reprogrammed” to hundred different types of cells that form accept a stem cell-like state (6). These tissues and organs and provide all the specialized cells, which are now called essential functions for survival and induced pluripotent stem cells (iPSCs), , reproduction (1). All of cells in the body, were reprogrammed to an embryonic stem both somatic (of all three germ layers) and cell-like state via introducing important germ cells, originate from the embryonic genes to maintaining the critical properties pluripotent cells (2). In 1961, for the first of embryonic stem cells (ESCs) (7). After time, Till and McCulloh (3) introduced this discovery, researchers have improved the generation techniques of iPSCs and Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] self-renewal properties in any living cells as one of their important breakthrough. created powerful new pathways to de- They influenced the mice with lethal differentiate cells, which their growth and radiation doses and then injected bone developmental conditions was already marrow cells in mice. They found that the determined (8-10). The main focus of main reason of survived of the mice was worldwide investigators and clinicians is these cells formed clumps due to cell on the potential use of iPSCs as a helpful cloned from them (4, 5). In 2006, Japanese tool for disease modeling, drug researchers at Kyoto University recognized development, and regenerative medicine Hematopoietic cancer therapy with induced pluripotent stem cells

(11-13). Furthermore, the ethical issues from somatic tissues of the rat and monkey related to the ESCs generation are not (27-29). applied to iPSCs, proposing a non- Several methods have been considered for controversial strategy to create pluripotent improving the effectiveness of the cells using existing non-pluripotent cell reprogramming and reducing the lines which can be transplanted into the potentially harmful side effects of the patients without the concern of immune reprogramming process (30). Since the rejection (autologous graft) (14, 15). used to deliver the four However, there are many challenges in transcription factors in the earliest studies pre-temporary iPSC tests, one of which is can be potentially mutagens, researchers development or tumorigenesis are not certain about whether all four risk if they are inserted in sensitive sites factors are absolutely essential (23, 31, such as promoters and host gene enhancers 32). Specially, the c- gene is known to (16). increase the rate of tumor growth in some cases, which negatively affects iPSC Experimental strategies for iPSCs efficiency in transplantation treatments generation (27, 33). For this purpose, researchers One strategy to achieve iPSCs generation proposed a three-factor approach using is nuclear reprogramming, a stable Oct4 with the orphan nuclear receptor alteration in the nucleus of a mature cell, Esrrb and , and they were able to which can then be preserved and replicated transform mouse embryonic as the cell divides through mitosis (17). into iPSCs. Similarly, other reports have This strategy is implemented using indicated that c-Myc has not a functional techniques such as fuse somatic cells with role for direct reprogramming of mouse ESCs, nuclear transfer fibroblasts (34, 35). Studies have further (SCNT), and altered nuclear transfer reduced the number of essential genes for (ANT) (18, 19). The nuclear reprogramming, and investigators continue reprogramming approach involves the use to recognize of chemicals that can replace of mature “somatic” cells from an adult or increase the efficiency of transcription and the introduction of genes that encoding factors in this procedure (36, 37). These critical proteins that successes continue to inform and facilitate themselves regulate the function of other the reprogramming process, thus important genes in the early developmental advancing the field toward the production stages of the fetus (20, 21). of patient specific stem cells for clinical In the previous studies, it was found that application. However, the way through only four transcription factors (Oct4: which transcription factors transfer to the Octamer binding transcription factor-4, somatic cells is critical to their potential Sox2: Sex determining region Y-box2, use in the clinic (38). : Krupple like factor 4, and c-Myc) were required for reprogramming of IPSCs in cancer therapy: mouse fibroblasts to an embryonic stem opportunities and challenges Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] cell–like state by forcing them to express Normally derived iPSCs from patient important genes for maintenance of the tissues can mainly be employed for tumor defining properties of ESCs (22-24). In regeneration or treatment of the affected 2007, two different research teams reached tissues. In regenerative medicine, a new milestone through deriving iPSCs numerous tissues can be produced by from human cells and using the original using iPSCs (2, 39). IPSCs therapy may be four genes containing Oct4, Sox2, Nanog, a good alternative for damaging and Lin28 (25, 26). Since then, researchers approaches such as chemotherapy, have achieved the production of iPSCs radiotherapy, or surgical treatment.

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 118

Khandany et al

However, regenerative therapy mediated researchers paid special attention to the by human iPSCs needs strong in vivo pluripotent stem cells that might be highly evidence from iPSC-derived tissues (19, tumorigenic. However, recently it is shown 40). that iPSCs is safer for clinical use than Now, only a few types of human iPSC- ESCs and other stem cells (49). However, derived cells (e.g. ) have been various strategies are used to minimize the effectively assessed in vivo (41, 42). In risk and possibility of neoplastic addition to the direct role of iPSCs in transformation. cancer treatment, they can be utilized to In the first step, undifferentiated screen novel anticancer drugs. Derived pluripotent stem cells, which are iPSCs from differentiating cancer tissue potentially tumorigenic, can be separated generate cell types that may be more from clinical approaches using biologically linked to human tumors and that focus on specific surface-displayed be more suitable for drug screening biomarkers. Stem cell differentiation methods. IPSCs are also better candidates downregulates expression of these than other stem cells for evaluation of the biomarkers (50). toxicities of antitumor drugs (25, 37, 43). Second, of iPSCs Similar to most chemotherapy involves monitoring of the expression of components, stem cell therapy using a the specific differentiation genes. single agent commonly cannot eliminate Successfully differentiated cells can be tumors. Therefore, a desired drug recognized and screened by using combination should be rationally generated recombinant reporter proteins. Green and selected (44, 45). Many of these fluorescent protein (GFP) and similar combination therapies have been tested to proteins work well as reporters of improve treatment durability. For example, differentiated against undifferentiated chemotherapy combined with interferon cells (51). Third, -guided toxins or (IFN)-beta immunotherapy, by using a toxic antibodies can kill the pro-drug/suicide gene system, has undifferentiated pluripotent stem cells displayed synergistic therapeutic effects in through immune pathways. For instance, human colorectal cancer (44, 46). In 2011, monoclonal antibodies against claudin-6, a Zielske et al., found that irradiating tumor surface biomarker for undifferentiated cells could induce production of factors pluripotent ESCs and iPSCs, can guide that stimulate stem cell invasion and immune-toxins to these stem cells for increase the number of stem cell in tumors targeted and selective killing (52). (47). Fourth, undifferentiated pluripotent stem There are three possibilities for tumors in cells can be removed from cytotoxic which cancer stem cells play a critical role. components, which can be utilized to kill First, the mutations in the normal stem targeted and selectively pluripotent stem cells or progenitor cells and transformation cells that can progress into tumors (53). of them into cancerous stem cells which Fifth, potentially tumorigenic pluripotent can lead to the growth of the primary stem cells can be transformed with suicide

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] tumor. Second, many of the primary tumor genes for sensitization towards pro-drugs. cells may be killed during chemotherapy, For this purpose, the enzyme/pro-drug but if the cancerous stem cells are not cancer therapy strategies can be improved eliminated, they become cancer-resistant to kill undifferentiated pluripotent stem stem cells and may result in tumor cells (54) Finally, tumorigenic pluripotent recurrence. Third, the cancer stem cells stem cells can be eradicated through self- may emigrate to distal positions from the induced transgenic expression of primary tumor and cause metastasis (48). recombinant human DNases (55). These For these reasons, most previous

119 Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Hematopoietic cancer therapy with induced pluripotent stem cells

strategies can protect a large population of patient (64). Kim et al., reported stem cells against tumor transformation. generation of single iPSC-like line in the parental pancreatic ductal adenocarcinoma Application of iPSCs in (PDAC) cancer cultures that contained regenerative medicine for cancer KRAS G12D mutation (65). The rarity of therapy successful studies demonstrates the difficulty of reprogramming primary It is proven that reprogramming of human cancer cells to iPSCs. However, technical primary cancer cells is very difficult. limitations, such as complications in However, there have been relatively few maintenance primary cancer tissues in reports demonstrating iPSCs production culture, cannot be excluded; furthermore, for successful reprogramming of tumoric the basic biological barriers may directly human cells. The generation of novel iPSC undermine the reprogramming process in and reprogramming cancer cells are cancer cells. thought to be faced with difficulties such In addition to particular characteristic of as reprogramming-triggered cellular pluripotent stem cells, such as their self- senescence, tumor-specific genetic renewal and differentiation capabilities, mutations, epigenetic modifications, they can repair human tissues after genomic instability, and accumulation of chemotherapy (66). Clinically, after DNA damage. In spite of these difficulties, treatment of malignancies with high-dose several researchers have reported the radiotherapy or chemotherapy, generation of new iPSC lines from present transplanting human stem cells has been cancer cell lines. Reports of iPSC lines generally used to facilitate lifelong resulting from human cancer cell lines are hematological recovery (67). The aim of summarized in Table I, which presents a this treatment is to reconstitute the bone range of cancers such as leukemia (AML) marrow under conditions of marrow (56), breast cancer (57), retinoblastoma failure (for example, in aplastic anemia or (58), colon cancer (44), Lliver cancer (59), other blood cell genetic diseases) and to hematologic and solid malignancies (60), apply human stem cells that differentiate melanoma (61), myelomonocytic leukemia into a desired type of hematopoietic cells (62), and glioblastoma (63). Based on in recipients (68). Normally originated these studies, reprogramming of cells with iPSCs from patient tissues can human cancer cell lines is not an theoretically be utilized to regenerate impossible task. However, certain types of tumor- or treatment- damaged tissues (69). cancers may be commonly used against In regenerative medicine approaches, reprogramming factors and the different tissues can be produced using combination of factors should be carefully iPSCs. IPSC therapy in regenerative selected depending on the type of cancer medicine may be valuable in replacing or cells that are attempting to reprogram. repairing cancer damaged cell by Nevertheless, reports of iPSCs generated radiotherapy, chemotherapy, or surgical from human primary malignant cells are treatment (70). However, regenerative few and limited to certain cancers, Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] therapy mediated by human iPSCs needs especially leukemia. Hu et al., were in vivo extensive studies in iPSC-derived successful in reprogramming primary tissues. So far, only a small number of human lymphoblasts using transgene-free human iPSC-derived cells, such as iPSC technology to ectopically express hepatocytes, have been successfully OSKM, LIN28, NANOG, and the SV40 evaluated in vivo (71, 72). large T gene from a BCR–ABL+ CML

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 120

Khandany et al

Table I: Applications of iPSCs in cancer therapy Origin cells used for Reprogramming factors Cancer type Type of therapy generation of iPSC Hematopoietic cells OSKM Leukemia investigating the mechanistic basis (Primary AML cells) and clonal properties of human AML, leukemic DNA methylation/gene expression patterns BRCA1-deleted blood OSKM breast cancer Cancer development modeling cells

Skin cells from RB OSNL Retinoblastoma Cancer development modeling patient

Familial OSKM Colon cancer Modeling of drug Testing in adenomatous Colorectal Cancer polyposis (Colonic )

Hepatic cells OSKM Liver cancer The engraft the liver in a mouse transplantation model

Natural killer cells (BJ1- OKSM hematologic Anti-tumor therapy (tumor targeting) iPS12, UCBiPS7, and and solid DRiPS16) malignancies

human tumor antigen- OSKM melanoma Anti-tumor therapy(tumor targeting) specific T cells (CD8+ mature T-cells)

Dendritic cells (cell lines: OSKM, BMP4 immunological Cancer immunotherapy such as DC- 201B7, 253G4, disorders based vaccines CIRA188Ai-W2, and CB- A11)

Juvenile Myelomonocytic OKSM Myelomonocyti Cancer development modeling Leukemia (JMML) c Leukemia

glioblastoma (neural OSKM, Oct4 glioblastoma Cancer development modeling lineage)

enucleated oocyte, and applying the IPSCs and hematopoietic cancer expression of specific reprogramming Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] therapy genes in somatic cells (74, 75). These two methods are used to reprogram multiple Developments in nuclear reprogramming various cell types, including pancreatic β of somatic cells lead to new ways of cells, liver cells, fibroblasts, T and B creating pluripotent stem cell lines and cells, and neural progenitor represent a significant step in the cells (76-78). Although many types of production of stem cell-based specific cells can be under nuclear reprogramming, therapies (73). Pluripotency the fundamental differences between reprogramming approach is based on pluripotent stem cell lines derived from transfer of a somatic cell nucleus into an

121 Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Hematopoietic cancer therapy with induced pluripotent stem cells

distinct somatic cell types are largely researches of cancer immunotherapy (88- unclear. 90). Patient-specific iPSCs can be also An immune-mediated antitumor effect potentially helpful in immunotherapy resulting from allogeneic hematopoietic methods (91, 92). The pre-rearranged TCR stem cell (HSC) transplantation might be gene is reserved in T lymphocyte-derived appropriate for the treatment of some human iPSCs, which can be further hematological malignancies, such as CML, differentiated into functionally active T ALL, AML, Hodgkin and Non-Hodgkin cells (93, 94). lymphoma, multiple myeloma (MM), In recent years, various technologies, sickle cell anemia, thalassemia, and generally based on reversing Fanconi anemia (79-81). These HSCs are immunosuppression, have been expanded the most common multipotent stem cells by utilizing transferring cytotoxic T that can differentiate to all blood cell lines lymphocyte (CTL) (95) and transferring through hematopoiesis process (82, 83). TCR gene (96). It is also necessary that After Yamanaka studies in the field of they differentiate into double positive cells reprogramming, which led invention of (CD4/8+), before generating iPSC-derived iPS technology, many researchers have CTL (iCTL), by IL-2 which is the major tried to generate induced hematopoietic in T-cell differentiation (97, 98). stem cells (iHSC) derived from patients Experimentally tumor antigen-specific T with blood malignancy (84, 85). can be generated by Additionally, these researchers have reprogramming designated T cells into shown that the specific expression of 13 iPSCs which are then differentiate back transcription factors such as OCT4 of into T lymphocytes for infusion in OKSM factors can generate HSC. patients. However, the safety of - Recently, it has been proven that new derived human iPSCs must be more genetic engineering tools can be used for credible (99, 100). producing of iHSCs, including CRISPR- Since most cancers in a specific tissue , meganucleases, zinc-finger include acquired nucleotide mutations, nucleases (ZFN), double-strand break iPSCs resulted from other normal tissues (DSB) nucleases, and transcription of the same patient hypothetically can be activator-like effector nucleases (TALEN) used to regenerate those injured tissues by (86, 87). Studies have revealed that two the tumors themselves or subsequent transcription factors (HOXA and ERG) act treatments (101, 102). However, as the core proliferation and differentiation applications the iPSC-derived tissues in factors in HSC and are inducers of the self- cancer therapy, needs that confiremed by renewal ability, and thus dysregulation of in vivo studies. It has been made clear that these factors progresses several types of human iPSCs derived from T lymphocytes leukemic cells (85). The main differential maintain the pre-rearranged T cell receptor marker of hematopoietic stem cells is (TCR) gene; therefore, these iPSCs can be CD34 that help distinguish iHSC in induced for differentiation into association with other differentiation functionally active T cells (93, 94). In

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] markers such as CD34, CD38, CD45, 2018, Chao et al., described the generation CD90, CD105, CD133, and C-kit (stem of AML-iPSCs from two patients, with cell receptor) (83). more emphasis on the notion that reprogramming of primary leukemias was 1- Reprogrammed T-lymphocytes not an unimportant assignment (103). Presenting genes encoding chimeric Nevertheless, natural or technical antigen receptors (CARs) or T-cell reprogramming difficulties are significant receptors (TCRs) directed against tumor- barriers to reprogramming of primary related antigens makes HSCs attractive for

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 122

Khandany et al

acute leukemic cells that need further able to produce large numbers of UCB investigation. CD34+ cells-derived NK-cells for adoptive The differences in reprogramming pre- immunotherapy in large-scale bioreactors, rearranged cells in AML and B-ALL and for the use in future clinical cancer propose that epigenetic reprogramming trials (116, 117). According to the studies may be dependent on the specific genetic on induced NK cells based on iPSC (iNK alterations and leukemia tumor subtype cells), reprogrammed NK cell can supply (104-106). Recent studies have shown that the desired volume of cell product to be dendritic cells derived from human iPSCs used in leukemia therapy panel without can be completely functional for the any immunological response. assessment of hematopoietic diseases (107-109). However, these cells still need Conclusion in vivo testing to confirm the effectiveness Induced pluripotent stem cells provide an and safety of their application. alternative approach to human embryonic stem cells (hESCs) without any ethical 2- Induced NK Cells concerns and with universal usage. Since Human natural killer (NK) cells are they are obtained from dedifferentiation of considered as an significant part of the adult cells and not from , iPSCs innate by producing overcome the ethical hurdles of embryonic significant for killing virally stem cells. There are still significant infected and/or malignant cells (110). obstacles that need to be considered. While antitumor T‐cell immune responses Despite the attractive research on iPSC- involve a diagnostic initiating phase and derived cell transplantation in the grafts, their responses are restricted to human there is still a potential risk of tumor leukocyte antigen (HLA), mature NK cells formation. It is possible to generate patient effectively kill malignant cells without any specific iPSC from somatic tissues, which previous exposure (111). In addition, NK can contribute to drug development, cells are the link between innate and disease modeling, and autologous stem adaptive immune systems that can increase cell therapy. Stem cells migrate to solid and accelerate immunologic responses tumors and facilitate site-specific anti- using their memory ability. At present, tumor drug delivery. It is clear that these cells are extracted by leukapheresis induced hematopoietic stem cells -derived from the blood circulation and then immune cells provide a genetic separated from other cells via magnetic manageable system to study human beads coated with anti-CD56. Both NK immune cell function and progress. In cell ‐ and T cell ‐based adoptive addition, these immune cells provide a immunotherapies have been used to treat main source of lymphocytes and may open patients with tumor malignancies. In recent new doors for cancer therapy. Finally, + studies, CD34 hematopoietic progenitor these studies have demonstrated that cell cells derived from umbilical cord blood reprogramming in primary tumor cells is (UCB) are also being used as a basis for more difficult, and that further Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] the generation of many numbers of technological development is needed to be allogeneic NK cells (112). Some research able to generate reliable iPSC models of groups have defined various protocols for cancers. + the production of NK cells from CD34 cells using culture of stromal cell lines and Conflicts of interest a combination of cytokines that progress There are no conflicts of interest. the expansion of NK cells (113-115). Furthermore, other researchers have been

123 Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Hematopoietic cancer therapy with induced pluripotent stem cells

References 12. Beyene R, Boockvar JA. Disease- 1. Colman A. Induced pluripotent specific induced pluripotent stem cells. stem cells and human disease. Cell Stem Neurosurgery 2008;63(6):12-18. Cell 2008;3(3):236-237. 13. Dimos JT, Rodolfa KT, Niakan 2. Xia X, Chu J, Chen X. Induced KK, Weisenthal LM, Mitsumoto H, Chung pluripotent stem cells generated from W, et al. Induced pluripotent stem cells reprogramming differentiated cells by generated from patients with ALS can be defined factors. Sheng Wu Gong Cheng differentiated into motor neurons. Science Xue Bao 2008 ;24(7):1121-1127. 2008;321(5893):1218-1221. 3. Till JE, Mc CE. A direct 14. Zhu D, Kong CSL, Gingold JA, measurement of the radiation sensitivity of Zhao R, Lee DF. Induced Pluripotent Stem normal mouse cells. Radiat Cells and Induced Pluripotent Cancer Cells Res 1961;14:213-222. in Cancer Disease Modeling. Adv Exp 4. Osawa M, Nakamura K, Nishi N, Med Biol 2018;1119:169-183 Takahasi N, Tokuomoto Y, Inoue H, et al. 15. Yu J, Vodyanik MA, Smuga-Otto In vivo self-renewal of c-Kit+ Sca-1+ K, Antosiewicz-Bourget J, Frane JL, Tian Lin(low/-) hemopoietic stem cells. J S, et al. Induced pluripotent stem cell lines Immunol 1996;156(9):3207-3214. derived from human somatic cells. Science 5. Spangrude GJ, Heimfeld S, 2007;318(5858):1917-1920. Weissman IL. Purification and 16. Duinsbergen D, Salvatori D, characterization of mouse hematopoietic Eriksson M, Mikkers H. Tumors stem cells. Science 1988;241(4861):58-62. originating from induced pluripotent stem 6. Okita K, Ichisaka T, Yamanaka S. cells and methods for their prevention. Generation of germline-competent induced Ann N Y Acad Sci 2009;1176:197-204. pluripotent stem cells. Nature 17. Yuan TF, Arias-Carrion O. Locally 2007;448(7151):313-317. induced neural stem cells/pluripotent stem 7. Bindhya S, Sidhanth C, Shabna A, cells for in vivo cell replacement therapy. Krishnapriya S, M G, Ganesan TS. Int Arch Med 2008;1(1):17-20. Induced Pluripotent Stem Cells: A New 18. Yaddanapudi K, Li C, Eaton JW. Strategy to Model Human Cancer. Int J with induced pluripotent stem Biochem Cell Biol 2019;107:62-68. cells confers protection against cancer. 8. Camara DA, Mambelli LI, Stem Cell Investig 2018;5:23-28. Porcacchia AS, Kerkis I. Advances and 19. Tulloch NL, Pabon L, Murry CE. Challenges on Cancer Cells Get with the (re)program: cardiovascular Reprogramming Using Induced potential of skin-derived induced Pluripotent Stem Cells Technologies. J pluripotent stem cells. Circulation Cancer 2016;7(15):2296-2303. 2008;118(5):472-475. 9. Chamberlain SJ, Li XJ, Lalande M. 20. Stadtfeld M, Nagaya M, Utikal J, Induced pluripotent stem (iPS) cells as in Weir G, Hochedlinger K. Induced vitro models of human neurogenetic pluripotent stem cells generated without disorders. Neurogenetics 2008;9(4):227- viral integration. Science Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] 235. 2008;322(5903):945-949. 10. Durcova-Hills G. Induced 21. Shi Y, Do JT, Desponts C, Hahm reprogramming of human somatic cells HS, Scholer HR, Ding S. A combined into pluripotency: a new way how to chemical and genetic approach for the generate pluripotent stem cells. generation of induced pluripotent stem Differentiation 2008;76(4):323-325. cells. Cell Stem Cell 2008;2(6):525-528. 11. Aoi T. Advance in study of 22. Sato M, Kawana K, Adachi K, induced pluripotent stem cells (iPS cells). Fujimoto A, Yoshida M, Nakamura H, et Nihon Rinsho 2008;66(5):850-856. al. Regeneration of cervical reserve cell-

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 124

Khandany et al

like cells from human induced pluripotent induced pluripotent stem cells for research stem cells (iPSCs): A new approach to and therapy. Nat Rev Mol Cell Biol finding targets for cervical 2008;9(9):725-729. treatment. Oncotarget 2017;8(25):40935- 33. Zhang DM, Li JJ, Yan P, Hu JT. 40945. Establishment and identification of 23. Park IH, Lerou PH, Zhao R, Huo induced pluripotent stem cells in liver H, Daley GQ. Generation of human- cancer patients. Asian Pac J Trop Med induced pluripotent stem cells. Nat Protoc 2014;7(4):253-256. 2008;3(7):1180-1186. 34. Chen L, Kasai T, Li Y, Sugii Y, Jin 24. Singhal DK, Singhal R, Malik HN, G, Okada M, et al. A model of cancer stem Singh S, Kumar S, Kaushik JK, et al. cells derived from mouse induced Molecular cloning and production of pluripotent stem cells. PLoS One caprine recombinant Oct4 protein for 2012;7(4):e33544-e33450. generation induced pluripotent stem cells. 35. Geoghegan E, Byrnes L. Mouse Mol Biol Rep 2015;42(12):1583-1591. induced pluripotent stem cells. Int J Dev 25. Blelloch R, Venere M, Yen J, Biol 2008;52(8):1015-1022. Ramalho-Santos M. Generation of induced 36. Aasen T, Raya A, Barrero MJ, pluripotent stem cells in the absence of Garreta E, Consiglio A, Gonzalez F, et al. drug selection. Cell Stem Cell Efficient and rapid generation of induced 2007;1(3):245-247. pluripotent stem cells from human 26. Kim JB, Zaehres H, Wu G, Gentile keratinocytes. Nat Biotechnol L, Ko K, Sebastiano V, et al. Pluripotent 2008;26(11):1276-1284. stem cells induced from adult neural stem 37. Anguera MC, Sadreyev R, Zhang cells by reprogramming with two factors. Z, Szanto A, Payer B, Sheridan SD, et al. Nature 2008;454(7204):646-650. Molecular signatures of human induced 27. Nakagawa M, Koyanagi M, Tanabe pluripotent stem cells highlight sex K, Takahashi K, Ichisaka T, Aoi T, et al. differences and cancer genes. Cell Stem Generation of induced pluripotent stem Cell 2012;11(1):75-90. cells without Myc from mouse and human 38. Eminli S, Utikal J, Arnold K, fibroblasts. Nat Biotechnol Jaenisch R, Hochedlinger K. 2008;26(1):101-106. Reprogramming of neural progenitor cells 28. Liu H, Zhu F, Yong J, Zhang P, into induced pluripotent stem cells in the Hou P, Li H, et al. Generation of induced absence of exogenous Sox2 expression. pluripotent stem cells from adult rhesus Stem Cells 2008;26(10):2467-2474. monkey fibroblasts. Cell Stem Cell 39. Yang J, Lam DH, Goh SS, Lee EX, 2008;3(6):587-590. Zhao Y, Tay FC, et al. Tumor tropism of 29. Liao J, Cui C, Chen S, Ren J, Chen intravenously injected human-induced J, Gao Y, et al. Generation of induced pluripotent stem cell-derived neural stem pluripotent stem cell lines from adult rat cells and their gene therapy application in cells. Cell Stem Cell 2009;4(1):11-15. a metastatic breast cancer model. Stem 30. Singh VK, Kumar N, Kalsan M, Cells 2012;30(5):1021-1029.

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] Saini A, Chandra R. Mechanism of 40. Wartenberg M, Donmez F, Ling Induction: Induced Pluripotent Stem Cells FC, Acker H, Hescheler J, Sauer H. (iPSCs). J Stem Cells 2015;10(1):43-62. Tumor-induced angiogenesis studied in 31. Park IH, Arora N, Huo H, Maherali confrontation cultures of multicellular N, Ahfeldt T, Shimamura A, et al. tumor spheroids and embryoid bodies Disease-specific induced pluripotent stem grown from pluripotent embryonic stem cells. Cell 2008;134(5):877-886. cells. FASEB J 2001;15(6):995-1005. 32. Nishikawa S, Goldstein RA, Nierras CR. The promise of human

125 Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Hematopoietic cancer therapy with induced pluripotent stem cells

41. Chen L, Mizutani A, Kasai T, Yan Regulation of Epidermal Stem Cell T, Jin G, Vaidyanath A, et al. Mouse Biomarkers and Their Role in Wound induced pluripotent stem cell Healing. Int J Mol Sci 2015;17(1):e16-e20. microenvironment generates epithelial- 51. Luo Y, Liu C, Cerbini T, San H, mesenchymal transition in mouse Lewis Lin Y, Chen G, et al. Stable enhanced cancer cells. Am J Cancer Res green fluorescent protein expression after 2014;4(1):80-88. differentiation and transplantation of 42. Kim H, Schaniel C. Modeling reporter human induced pluripotent stem Hematological Diseases and Cancer With cells generated by AAVS1 transcription Patient-Specific Induced Pluripotent Stem activator-like effector nucleases. Stem Cells. Front Immunol 2018;9:2243-2248. Cells Transl Med 2014;3(7):821-835. 43. Yang M, Liu Y, Hou W, Zhi X, 52. Zhang CL, Huang T, Wu BL, He Zhang C, Jiang X, et al. Mitomycin C- WX, Liu D. Stem cells in cancer therapy: treated human-induced pluripotent stem opportunities and challenges. Oncotarget cells as a safe delivery system of gold 2017;8(43):75756-75766. nanorods for targeted photothermal 53. Schriebl K, Satianegara G, Hwang therapy of gastric cancer. Nanoscale A, Tan HL, Fong WJ, Yang HH, et al. 2017;9(1):334-340. Selective removal of undifferentiated 44. Crespo M, Vilar E, Tsai SY, Chang human embryonic stem cells using K, Amin S, Srinivasan T, et al. magnetic activated cell sorting followed by Corrigendum: Colonic organoids derived a cytotoxic antibody. Tissue Eng Part A from human induced pluripotent stem cells 2012;18(9-10):899-909. for modeling colorectal cancer and drug 54. Xu G, McLeod HL. Strategies for testing. Nat Med 2018;24(4):526-531. enzyme/prodrug cancer therapy. Clin 45. Fernandez Tde S, De Souza Cancer Res 2001;7(11):3314-3324. Fernandez C, Mencalha AL. Human 55. Malecki M, LaVanne C, Alhambra induced pluripotent stem cells from basic D, Dodivenaka C, Nagel S, Malecki R. research to potential clinical applications Safeguarding Stem Cell-Based in cancer. Biomed Res Int Regenerative Therapy against Iatrogenic 2013;2013:430290-430299. Cancerogenesis: Transgenic Expression of 46. Marin Navarro A, Susanto E, Falk DNASE1, DNASE1L3, DNASE2, DFFB A, Wilhelm M. Modeling cancer using Controlled By POLA1 Promoter in patient-derived induced pluripotent stem Proliferating and Directed Differentiation cells to understand development of Resisting Human Autologous Pluripotent childhood malignancies. Cell Death Induced Stem Cells Leads to their Death. J Discov 2018;4:7-10. Stem Cell Res Ther 2013; 9(5): 21559- 47. Zielske SP, Spalding AC, Wicha 21567. MS, Lawrence TS. Ablation of breast 56. Chao MP, Gentles AJ, Chatterjee cancer stem cells with radiation. Transl S, Lan F, Reinisch A, Corces MR, et al. Oncol 2011;4(4):227-233. Human AML-iPSCs Reacquire Leukemic 48. Hu Y, Fu L. Targeting cancer stem Properties after Differentiation and Model

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] cells: a new therapy to cure cancer Clonal Variation of Disease. Cell Stem patients. Am J Cancer Res 2012;2(3):340- Cell 2017;20(3):329-344. 356. 57. Griscelli F, Oudrhiri N, Feraud O, 49. Hirschi KK, Li S, Roy K. Induced Divers D, Portier L, Turhan AG, et al. pluripotent stem cells for regenerative Generation of induced pluripotent stem medicine. Annu Rev Biomed Eng cell (iPSC) line from a patient with triple 2014;16:277-294. negative breast cancer with hereditary 50. Saldanha SN, Royston KJ, exon 17 deletion of BRCA1 gene. Stem Udayakumar N, Tollefsbol TO. Epigenetic Cell Res 2017;24:135-138.

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 126

Khandany et al

58. Zeng S, Liu L, Ouyang Q, Zhao Y, 66. Aponte PM, Caicedo A. Stemness Lin G, Hu L, et al. Generation of induced in Cancer: Stem Cells, Cancer Stem Cells, pluripotent stem cells (iPSCs) from a and Their Microenvironment. Stem Cells retinoblastoma patient carrying a Int 2017;2017:5619472-5619479. c.2663G>A mutation in RB1 gene. Stem 67. Van Zant G, Liang Y. Concise Cell Res 2016;17(2):208-211. review: aging, life 59. Liu H, Kim Y, Sharkis S, span, and transplantation. Stem Cells Marchionni L, Jang YY. In vivo liver Transl Med 2012;1(9):651-657. regeneration potential of human induced 68. Bryder D, Rossi DJ, Weissman IL. pluripotent stem cells from diverse origins. Hematopoietic stem cells: the paradigmatic Sci Transl Med 2011;3(82):82-93. tissue-specific stem cell. Am J Pathol 60. Knorr DA, Ni Z, Hermanson D, 2006;169(2):338-346. Hexum MK, Bendzick L, Cooper LJ, et al. 69. Singh VK, Kalsan M, Kumar N, Clinical-scale derivation of natural killer Saini A, Chandra R. Induced pluripotent cells from human pluripotent stem cells for stem cells: applications in regenerative cancer therapy. Stem Cells Transl Med medicine, disease modeling, and drug 2013;2(4):274-283. discovery. Front Cell Dev Biol 2015;3:2-9. 61. Vizcardo R, Masuda K, Yamada D, 70. Sankar S, Sharma CS, Rath SN, Ikawa T, Shimizu K, Fujii S, et al. Ramakrishna S. Electrospun Fibers for Regeneration of human tumor antigen- Recruitment and Differentiation of Stem specific T cells from iPSCs derived from Cells in Regenerative Medicine. mature CD8(+) T cells. Cell Stem Cell Biotechnol J 2017;12(12): 1700263- 2013;12(1):31-36. 1700269. 62. Gagne AL, Maguire JA, Gandre- 71. Harding J, Mirochnitchenko O. Babbe S, Chou ST, Tasian SK, Loh ML, et Preclinical studies for induced pluripotent al. Generation of a human Juvenile stem cell-based therapeutics. J Biol Chem myelomonocytic leukemia iPSC line, 2014;289(8):4585-4593. CHOPi001-A, with a mutation in CBL. 72. Kondo Y, Toyoda T, Inagaki N, Stem Cell Res 2018;31:157-160. Osafune K. iPSC technology-based 63. Caren H, Stricker SH, Bulstrode H, regenerative therapy for . J Gagrica S, Johnstone E, Bartlett TE, et al. Diabetes Investig 2018;9(2):234-243. Glioblastoma Stem Cells Respond to 73. Patel M, Yang S. Advances in Differentiation Cues but Fail to Undergo reprogramming somatic cells to induced Commitment and Terminal Cell-Cycle pluripotent stem cells. Stem Cell Rev Arrest. Stem Cell Reports 2015;5(5):829- 2010;6(3):367-380. 842. 74. Zuo Y, Su G, Cheng L, Liu K, 64. Hu K, Yu J, Suknuntha K, Tian S, Feng Y, Wei Z, et al. Coexpression Montgomery K, Choi KD, et al. Efficient analysis identifies nuclear reprogramming generation of transgene-free induced barriers of somatic cell nuclear transfer pluripotent stem cells from normal and . Oncotarget 2017;8(39):65847- neoplastic bone marrow and cord blood 65859.

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] mononuclear cells. Blood 75. Tian XC, Kubota C, Enright B, 2011;117(14):e109-119. Yang X. Cloning animals by somatic cell 65. Kim J, Hoffman JP, Alpaugh RK, nuclear transfer--biological factors. Reprod Rhim AD, Reichert M, Stanger BZ, et al. Biol Endocrinol 2003;1:98-100. An iPSC line from human pancreatic 76. Mall M, Wernig M. The novel tool ductal adenocarcinoma undergoes early to of cell reprogramming for applications in invasive stages of pancreatic cancer molecular medicine. J Mol Med (Berl) progression. Cell Rep 2013;3(6):2088- 2017;95(7):695-703. 2099.

127 Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Hematopoietic cancer therapy with induced pluripotent stem cells

77. Mayhew CN, Wells JM. 87. Hotta A, Yamanaka S. From Converting human pluripotent stem cells Genomics to Gene Therapy: Induced into beta-cells: recent advances and future Pluripotent Stem Cells Meet Genome challenges. Curr Opin Organ Transplant Editing. Annu Rev Genet 2015;49:47-70. 2010;15(1):54-60. 88. Larson S, De Oliveira SN. Gene- 78. Kawamata M, Suzuki A. Cell fate modified hematopoietic stem cells for modification toward the hepatic lineage by cancer immunotherapy. Hum Vaccin extrinsic factors. J Biochem Immunother 2014;10(4):982-985. 2017;162(1):11-16. 89. Gschweng E, De Oliveira S, Kohn 79. Renga M, Pedrazzoli P, Siena S. DB. Hematopoietic stem cells for cancer Present results and perspectives of immunotherapy. Immunol Rev allogeneic non-myeloablative 2014;257(1):237-249. hematopoietic stem cell transplantation for 90. Barrett DM, Grupp SA, June CH. treatment of human solid tumors. Ann Chimeric Antigen Receptor- and TCR- Oncol 2003;14(8):1177-1184. Modified T Cells Enter Main Street and 80. Karadurmus N, Sahin U, Bahadir Wall Street. J Immunol 2015;195(3):755- Basgoz B, Arpaci F, Demirer T. A Review 761. of Allogeneic Hematopoietic Stem Cell 91. Jiang Z, Han Y, Cao X. Induced Transplantation in Metastatic Breast pluripotent stem cell (iPSCs) and their Cancer. Int J Hematol Oncol Stem Cell application in immunotherapy. Cell Mol Res 2018;12(2):111-116. Immunol 2014;11(1):17-24. 81. Hatzimichael E, Tuthill M. 92. Jiang Y, Habibollah S, Tilgner K, Hematopoietic stem cell transplantation. Collin J, Barta T, Al-Aama JY, et al. An Stem Cells Cloning 2010;3:105-117. induced pluripotent stem cell model of 82. Birbrair A, Frenette PS. Niche hypoplastic left heart syndrome (HLHS) heterogeneity in the bone marrow. Ann N reveals multiple expression and functional Y Acad Sci 2016;1370(1):82-96. differences in HLHS-derived cardiac 83. Maali A, Atashi A, Ghaffari S, myocytes. Stem Cells Transl Med Kouchaki R, Abdolmaleki F, Azad M. A 2014;3(4):416-423. Review on Leukemia and iPSC 93. Vizcardo R, Klemen ND, Islam Technology: Application in Novel SMR, Gurusamy D, Tamaoki N, Yamada Treatment and Future. Curr Stem Cell Res D, et al. Generation of Tumor Antigen- Ther 2018;13(8):665-675. Specific iPSC-Derived Thymic Emigrants 84. Szabo E, Rampalli S, Risueno RM, Using a 3D Thymic Culture System. Cell Schnerch A, Mitchell R, Fiebig-Comyn A, Rep 2018;22(12):3175-3190. et al. Direct conversion of human 94. Saito H, Okita K, Chang AE, Ito F. fibroblasts to multilineage blood Adoptive Transfer of CD8+ T Cells progenitors. Nature 2010;468(7323):521- Generated from Induced Pluripotent Stem 526. Cells Triggers Regressions of Large 85. Doulatov S, Vo LT, Chou SS, Kim Tumors Along with Immunological PG, Arora N, Li H, et al. Induction of Memory. Cancer Res 2016;76(12):3473-

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] multipotential hematopoietic progenitors 3483. from human pluripotent stem cells via 95. Chapuis AG, Ragnarsson GB, respecification of lineage-restricted Nguyen HN, Chaney CN, Pufnock JS, precursors. Cell Stem Cell Schmitt TM, et al. Transferred WT1- 2013;13(4):459-470. reactive CD8+ T cells can mediate 86. Perez-Pinera P, Ousterout DG, antileukemic activity and persist in post- Gersbach CA. Advances in targeted transplant patients. Sci Transl Med genome editing. Curr Opin Chem Biol 2013;5(174):174-179. 2012;16(3-4):268-277.

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 128

Khandany et al

96. Robbins PF, Morgan RA, Feldman 105. McClellan JS, Dove C, Gentles AJ, SA, Yang JC, Sherry RM, Dudley ME, et Ryan CE, Majeti R. Reprogramming of al. Tumor regression in patients with primary human Philadelphia chromosome- metastatic synovial cell sarcoma and positive acute lymphoblastic melanoma using genetically engineered leukemia cells into nonleukemic lymphocytes reactive with NY-ESO-1. J . Proc Natl Acad Sci U S A Clin Oncol 2011;29(7):917-924. 2015;112(13):4074-4079. 97. Maeda T, Nagano S, Ichise H, 106. Wouters BJ, Delwel R. Kataoka K, Yamada D, Ogawa S, et al. and approaches to targeted epigenetic Regeneration of CD8alphabeta T Cells therapy in acute myeloid leukemia. Blood from T-cell-Derived iPSC Imparts Potent 2016;127(1):42-52. Tumor Antigen-Specific Cytotoxicity. 107. Li Y, Liu M, Yang ST. Dendritic Cancer Res 2016;76(23):6839-6850. cells derived from pluripotent stem cells: 98. Hoyer S, Prommersberger S, Potential of large scale production. World Pfeiffer IA, Schuler-Thurner B, Schuler G, J Stem Cells 2014;6(1):1-10. Dorrie J, et al. Concurrent interaction of 108. Vo LT, Daley GQ. De novo DCs with CD4(+) and CD8(+) T cells generation of HSCs from somatic and improves secondary CTL expansion: It pluripotent stem cell sources. Blood takes three to tango. Eur J Immunol 2015;125(17):2641-2648. 2014;44(12):3543-3559. 109. Iizuka-Koga M, Asashima H, Ando 99. Kaneko S. In Vitro Generation of M, Lai CY, Mochizuki S, Nakanishi M, et Antigen-Specific T Cells from Induced al. Functional Analysis of Dendritic Cells Pluripotent Stem Cells of Antigen-Specific Generated from T-iPSCs from CD4+ T T Cell Origin. Methods Mol Biol Cell Clones of Sjogren's Syndrome. Stem 2016;1393:67-73. Cell Reports 2017;8(5):1155-1163. 100. Brauer PM, Singh J, Xhiku S, 110. Paul S, Lal G. The Molecular Zuniga-Pflucker JC. T Cell Genesis: In Mechanism of Natural Killer Cells Vitro Veritas Est? Trends Immunol Function and Its Importance in Cancer 2016;37(12):889-901. Immunotherapy. Front Immunol 101. Papapetrou EP. Patient-derived 2017;8:1124-1128. induced pluripotent stem cells in cancer 111. Sharma P, Kumar P, Sharma R. research and precision oncology. Nat Med Natural Killer Cells - Their Role in 2016;22(12):1392-1401. Tumour Immunosurveillance. J Clin Diagn 102. Mahla RS. Stem Cells Applications Res 2017;11(8):BE01-BE05. in Regenerative Medicine and Disease 112. Spanholtz J, Preijers F, Tordoir M, Therapeutics. Int J Cell Biol Trilsbeek C, Paardekooper J, Witte T, et 2016;2016:6940283-6940288. al. Clinical-grade generation of active NK 103. Chao HM, Chern E. Patient- cells from cord blood hematopoietic derived induced pluripotent stem cells for progenitor cells for immunotherapy using models of cancer and cancer stem cell a closed-system culture process. PLoS One research. J Formos Med Assoc 2011;6(6):e20740-20745.

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ] 2018;117(12):1046-1057. 113. Luevano M, Madrigal A, 104. Munoz-Lopez A, Romero-Moya D, Saudemont A. Generation of natural killer Prieto C, Ramos-Mejia V, Agraz-Doblas cells from hematopoietic stem cells in vitro A, Varela I, et al. Development for immunotherapy. Cell Mol Immunol Refractoriness of MLL-Rearranged 2012;9(4):310-320. Human B Cell Acute Leukemias to 114. Cany J, Van der Waart AB, Reprogramming into Pluripotency. Stem Spanholtz J, Tordoir M, Jansen JH, Van Cell Reports 2016;7(4):602-618. der Voort R, et al. Combined IL-15 and IL-12 drives the generation of CD34(+)-

129 Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130

Hematopoietic cancer therapy with induced pluripotent stem cells

derived natural killer cells with superior maturation and alloreactivity potential following adoptive transfer. Oncoimmunology 2015;4(7):e1017701- e1017705. 115. Dezell SA, Ahn YO, Spanholtz J, Wang H, Weeres M, Jackson S, et al. Natural killer cell differentiation from hematopoietic stem cells: a comparative analysis of heparin- and stromal cell- supported methods. Biol Blood Marrow Transplant 2012;18(4):536-545. 116. Eguizabal C, Zenarruzabeitia O, Monge J, Santos S, Vesga MA, Maruri N, et al. Natural killer cells for cancer immunotherapy: pluripotent stem cells- derived NK cells as an immunotherapeutic perspective. Front Immunol 2014;5:439- 449. 117. Spanholtz J, Tordoir M, Eissens D, Preijers F, Van der Meer A, Joosten I, et al. High log-scale expansion of functional human natural killer cells from umbilical cord blood CD34-positive cells for adoptive cancer immunotherapy. PLoS One 2010;5(2):e9221-e9224.

Downloaded from ijpho.ssu.ac.ir at 5:39 IRST on Tuesday September 28th 2021 [ DOI: 10.18502/ijpho.v9i2.611 ]

Iran J Ped Hematol Oncol. 2019, Vol 9. No 2, 117-130 130