bentham.manuscriptpoint.com · web viewleukemia is an uncontrollable growth of hematopoietic cells...

29
A Review on Leukemia and iPS Technology: Application in Novel Treatment and Future Amirhosein Maali 1 , Amir Atashi 2 , Sasan Ghaffari 3 , Reza Kouchaki 1 , Fereshteh Abdolmaleki 1 , Mehdi Azad 1 * 1 Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran 2 Stem Cells and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran 3 Department of Hematology, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran *Corresponding Author: Mehdi Azad. PhD. Qazvin, Shahid Bahonar Boulevard, University of Medical Sciences, Faculty of Allied Medicine (Hematology), Department of Medical laboratory sciences. Tel: +982833359501 Fax: +982833338034 Zip code: 3419759811 Email Address: [email protected] 1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35

Upload: vohanh

Post on 08-Apr-2019

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

A Review on Leukemia and iPS Technology: Application in Novel Treatment and Future

Amirhosein Maali1, Amir Atashi2, Sasan Ghaffari3, Reza Kouchaki1, Fereshteh Abdolmaleki1, Mehdi Azad1*

1Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran2Stem Cells and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran3Department of Hematology, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran

*Corresponding Author: Mehdi Azad. PhD. Qazvin, Shahid Bahonar Boulevard, University of Medical Sciences, Faculty of Allied Medicine (Hematology), Department of Medical laboratory sciences. Tel: +982833359501 Fax: +982833338034 Zip code: 3419759811Email Address: [email protected]

1

12

3

456

7

89

1011121314151617

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

Page 2: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

Abstract

Leukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal disorder that generally leads to a clonal abnormality. Theoretical and technical inability in early screening and distinguishing cancer, tumor tolerance to common treatment methods, repeated relapses of cancer after remission phase, heterogeneous chromosomal abnormality, and the side effects of current chemotherapies are some challenges that we face with leukemia and other malignancies. Induced pluripotent stem cells (iPSC) opened a promising window to a wide range of diseases, including leukemia. Overcoming the barriers in leukemia is possible with iPSC technology. Induced hematopoietic stem cell transplantation (and gene therapy), induced cytotoxic T-lymphocytes and reprogrammed NK cells that strengthen the immune system, miRNAs, modeling approaches, and supportive cares are aspects of the novel treatment based on iPSC technology.

Keywords: Leukemia, iPS, treatment, Reprogrammed NK cells, miRNAs, gene therapy, iHSC, malignancy modeling

2

38

394041424344454647

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

Page 3: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

1. Introduction

Cancer is the uncontrollable cell growth, which is one of the leading causes of death worldwide. According to published statistics, cancer is the second and third reason for death in developed and developing countries, respectively.

Although in recent decades, significant developments in cancer diagnosis and treatment have been achieved, many areas in this field are still uncharted. Rudimentary knowledge and basic methods of cancer screening, imperfect treatment processes, and tumors tolerant to common treatments are all challenges that we face with a malignancy. Furthermore, repeated post-remission relapses, heterogeneous chromosomal abnormality, and malignant cells’ plasticity only serve to exacerbate the situation [1-3].

Cancer, almost, originates from one or more chromosomal disorder, which will generally lead to a clonal abnormality, a change in the gene expression profile, cell-cycle dysregulation, apoptosis, and ultimately incoherent cell growth. The genetic disorders that occur in intracellular key molecules and proto-oncogene and tumor suppressor genes result in malignancy. Since roughly 2004, the researchers’ efforts toward better understanding of cancer biology have evolved and improved the approach to prognosis, diagnosis, and treatment of cancer [4]. Naturally, chromosomal disorders arise before tumor forms in the body, and if they could be diagnosed before the tumor took place, it would be much easier to deal with and control cancer [5].

2. Review to Leukemia and Its Variants

Leukemia also occurs due to a mutation in DNA followed by interference in cellular function. The exact reason for each leukemia has not been recognized yet, but it is believed that its inception has environmental and inheritable reasons. There are four major classes of leukemia:

2.1. Acute Lymphoblastic Leukemia (ALL) is a relatively rare type of leukemia that leads to an increased number of WBCs and lymphoid cells in the peripheral blood and bone marrow (BM). Common translocations in ALL are Philadelphia (in 1.6% of patients; weak prognosis), t(4:11)(q21:q23) (in 1.6% of patients; weak prognosis), t(8:14)(q24:q32) (rare prevalence; weak prognosis), t(1:9)(q23:p13) (in 4.8% of patient), t(12:21) (in 25.4% of patients), t(11:14) (p13:q11) (rare prevalence). Other aberrancies include low-level hypodiploidy (weak prognosis), high-level hyperdiploidy (especially trisomy 4, 10, 17 with favorable prognosis), del(9p) (favorable prognosis), and complex karyotype (weak prognosis) [6-14].

Diagnosis of the affected lymphoid cell line plays a key role in choosing treatment protocol. Utilizing immunophenotyping assays can help to distinguish the type of ALL (prevalence of B-ALL and T-ALL is %80 and %20, respectively) [15, 16]. The B-ALL immunophenotypic profile is determined by specific B-cell CD markers, such as CD19, CD20, CD22, and CD79a. Primary B-cell blasts are generally CD10+ and CD34+, and more mature neoplastic cells express high levels of CD20. This can particularly be used to estimate the commencement of the disease. To identify the immunophenotypic profile of T-ALL, CD7, CD3, CD5, CD2, and CD1a can be used [17]. ALL in children has high incidence rate and the cure-rate (CR) is achieved in 75 to 90% of the patients by unique treatment methods [18-20].

2.2. Chronic lymphoblastic leukemia (CLL) is a type of leukemia that is accompanied by clonal proliferation and amassing of mature lymphocytes in peripheral blood, bone marrow, and lymph nodes [21]. Leukemic transformations in CLL are coupled with genetic changes and deletions of specific miRNA genes, developing survival properties of T and B-cell and resistance to apoptosis [22-24].

Long arm deletions of chromosome 13, especially 13q14 band (del (13q14)), are seen in more than 55% of cases. Recently, it has been shown that miR16-1 and miR15a are in the deleted areas (13q14) [22]. Other common aberrancies include long arm deletion of chromosome 11 (del (11q)) (in 25% of patients) [25, 26], trisomy 12 (in 10 to 20% of patients) [27], and short arm deletion of chromosome 17 (del (17p)) (in 5% of patients) [28].

3

71

72

737475

7677787980

81828384858687

88

89

909192

93949596979899

100101102103104105106107

108109110111

112113114115

Page 4: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

Primary diagnosis of CLL is accomplished by morphologically inspecting lymphocytes and witnessing a count of 5000 cells/µl of peripheral blood for at least 3 consecutive months. Most of the leukemic lymphocytes that can be seen in peripheral blood smear are tiny, mature cells with small cytoplasm and condensed nucleus. These cells may be present in combination with large and atypical cells, or with prolymphocytosis (ultimately up to 55% of the peripheral blood lymphoid cells). Lymph node agitation, lack of immunity, anemia, and coagulation disorders are the features of CLL [29]. In today’s society, this leukemia is widespread and is more prevalent in people over the age of 55. The 5-year survival rate in these individuals with advanced treatments is up to 75% [30].

2.3. Acute Myeloblastic Leukemia (AML) is followed by a heterogeneous disorder with clonal expansion of myeloid progenitor blasts in bone marrow and peripheral blood. The molecular screening has the main role in diagnosis, prognosis, and treatment process in AML. Genetic abnormality is seen in 50% of AML patients [31]. For example, the 5, 7, and 11q23 chromosomal changes and complex karyotype (simultaneous three-chromosomal abnormalities) are accompanied by lower response to treatment and bad prognosis, whereas abnormalities like t(15:17)(q22:q12), t(8:21)(q22:q22), and inv(16)(p13.1:q22) have more favorable prognosis [32, 33]. However, about 40 to 50% of AML patients are cytogenetically normal, named CN-AML (cytogenetically normal AML) [32]. Recently, utilizing gene sequencing to investigate mutations has been a powerful tool to analyze AML and treat AML patients. This disorder is reported more in children and men, respectively, than in adults and women [34]. Previously, AML had a very poor prognosis, but with the aid of improved treatment regimens and supportive cares, such as antibiotics and blood transfusion, AML has reached 35 to 40% CR for people under the age of 60 [33]. Indeed, the prognosis has been better in people over 60 years, but it is not sufficient.

2.4. Chronic Myeloblastic leukemia (CML) is a type of myeloproliferative disorder accompanied by a massive invasion of peripheral blood, bone marrow, and spleen by mature and immature myeloid cells. Currently, in more than 90% of cases, CML, which is known for a prominent increase in normal myeloid cells in both activity and function, is diagnosed in preliminary chronic phase (CML-CP). If CML is not treated, it will enter the accelerated phase and then the more aggressive blastic phase (BP). The most common genetic translocation in CML is reciprocal t(9:22)(q34:q11). Consequent recombinant protein, BCR-ABL1, has persistent tyrosine kinase activity that begets malignancy [35]. In-vitro studies have shown that BCR-ABL1 is an oncogene that causes the growth of malignant cells and suppression of apoptosis. Considering its importance, many CML treatments focus on this protein. This leukemia is seen more in adults, and up to 90% CR is reported [36].

3. iPS Technology: a Grand Medical Evolution

Stem cells are identified by two characteristics: the ability of self-renewal and plasticity (the differentiation potential) [37]. These abilities are the most important factors in cell therapy. Different plasticity potential of stem cells divides them into 5 groups, including totipotent, pluripotent, multipotent, oligopotent, and unipotent [38, 39].

Researchers deduced that pluripotency in pluripotent stem cells is influenced by a complex network of regulatory genes. In 2006, Takahashi and Yamanaka reached a new technology that allows the integration of transcription factors responsible for pluripotency making it possible to produce pluripotent embryonic cells by editing somatic cells’ genes. This process, that is the opposite of differentiation, is called reprogramming and the produced cells are named induced pluripotent stem cell (iPSC). In this project, human and mouse somatic cells return to embryonic state after receiving and subsequently expressing OCT4, KLF4, SOX2, and C-MYC (OKSM) transcription factors (Table 1) [40-42].

OCT

3/4

Oct

amer

bi

ndin

g tr

ansc

riptio

n fa

ctor

POU

cl

ass

hom

eobo

xes

and

pseu

doge

nes

6p21

.33

MYB

MYB

pr

oto-

onco

gene

tr

ansc

riptio

n fa

ctor

Myb

/SAN

T do

mai

n co

ntai

ning

6q23

.3

4

116117118119120121122

123124125126127128129130131132133134

135136137138139140141142143

144

145146147

148149150151152153154

Page 5: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

KLF4

Krup

pel-l

ike

fact

or 4

Krup

pel

like

fact

ors (

Zinc

fin

gers

C2

H2-t

ype)

9q31

.2

ERG

ERG

,E

TS

tran

scrip

tion

fact

or

ETS

tran

scrip

tion

fa

ctor

fa

mily

21q2

2.2

SOX2

Sex

dete

rmin

ing

regi

on Y

–bo

x 2 SR

Y

(sex

de

term

inin

g re

gion

Y)

bo

xes

3q26

.33

GATA

2

GATA

bin

ding

pr

otei

n 2

GATA

zin

c fin

ger

dom

ain

cont

aini

ng

3q21

.3

C-MYC

cellu

lar

mye

locy

tom

atos

is O

ncog

ene

(bH

LH

tran

scrip

tion

fact

or)

Basic

he

lix-lo

op-

helix

pro

tein

s

8q24

.21

GFI1b

Grow

th

fact

or

inde

pend

ent

1B

tr

ansc

riptio

n fa

ctor

Zinc

fin

gers

C2

H2-t

ype

SN

AG

tran

scrip

tiona

l re

pres

sors

9q3

4.13

NAN

OG

Nan

og

hom

ebox

NKL

sub

clas

s ho

meo

boxe

s an

d ps

eudo

gene

s

12p1

3.31

FOS

FOS

pr

oto-

onco

gene

Tran

scrip

tion

fact

or

subu

nit

Fos

tran

scrip

tion

fact

or fa

mily

Basic

leu

cine

zip

per

prot

eins

14q2

4.3

LIN28

Prot

ein

lin-2

8

Zinc

fin

gers

CC

HC-t

ype

1p36

.11

ETV6

ETV

varia

nt 6

ETS

tran

scrip

tion

fact

or fa

mily

12p1

3.2

HOXA

9

Hom

ebox

A9 HO

XL

subc

lass

ho

meb

oxes

7p15

.2

TBX2

1

T-bo

x 21

T-bo

xes

17q2

1.32

HOXA

5

Hom

ebox

A5

HOXL

su

bcla

ss

hom

eobo

xes

7p15

.2

RUNX3

Runt

rel

ated

tr

ansc

riptio

n fa

ctor

3

- 1p36

.11

RORA

RAR

rela

ted

orph

an

rece

ptor

A

Nuc

lear

ho

rmon

e re

cept

ors

15q2

2.2

GATA

3

GATA

bi

ndin

g pr

otei

n 3

GATA

zin

c fin

ger

dom

ain

cont

aini

ng

10p1

4

SOX4

Sex

dete

rmin

ing

regi

on Y

–bo

x 4 SR

Y

(sex

de

term

inin

g re

gion

Y)

-bo

xes

6p22

.3

IKZF1

IKAR

OS

fam

ily

zinc

finge

r 1

Prot

ein

phos

phat

ase

1 re

gula

tory

(s

ubun

its Z

inc

finge

rs C

2H2-

type

)

7p12

.2

Nam

e

Desc

riptio

n

Gene

fam

ily

Chro

mos

omal

lo

catio

n

Nam

e

Desc

riptio

n

Gene

fam

ily

Chro

mos

omal

lo

catio

n

5

Page 6: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

Table 1. Genes relevant to iPSC production and their chromosomal location. OKSM factors, NANOG, and LIN-28 factors have a part in pluripotency status. HOXA9, HOXA5, RORA, SOX4, ERG, GATA2, GFI1b, FOS, ETV8, and MYB are important in iHSC potency and GATA3, RUNX3, TBX21, and IKZF1 are involved in induced immunologic cells.

The constructed iPSC can differentiate into whole somatic cells. Transplanting differentiated cells can eliminate disease-causing cellular defects. These cells have also been utilized in disease modeling and drug discovery. The upside is there is no immunological rejection risk (autologous graft), no limitation in the number of required cells, and no ethical issues. The first vector used in cloning the OKSM factors was retroviral vector [43, 44]. There are many challenges in preclinical trials of iPSC, one being teratoma development. Integrating vectors can promote tumorigenesis if they are inserted in sensitive sites like promoters and enhancers of host gene [45]. One solution to overcome this challenge is non-integrating vectors and molecules [46-60]. Since making iPSC is a long-term process, in urgent situations, it is possible to use iPSC of a fully MHC-matched donor from stem cell banks, which are expanding every day [61].

In 2007, the first clinical trial in the field of iPSC-based therapy was conducted in treating sickle cell anemic mice by autologous transplantation [62]. Nevertheless, it is clear that there is a long way to reach human iPSC-based therapy, but many kinds of research in this field are being undertaken, and so we will discuss these studies in the following sections [47].

3.1. Hematopoietic stem cell transplantation and gene therapy based on iPSCs

Hematopoietic stem cell (HSC) or hematoblast is a multipotent stem cell that can differentiate to all blood cell lines through hematopoiesis [63]. Hematopoietic stem cell transplantation (HSCT) is the most advanced and effective method of leukemia treatment [64, 65]. HSCT can be used to treat malignant disorders, such as ALL, AML, CML, Hodgkin lymphoma (relapsed/refractory), Non-Hodgkin lymphoma, myelodysplastic syndrome, multiple myeloma (MM); and also nonmalignant disorders such as thalassemia, sickle cell anemia, Fanconi anemia, aplastic anemia, immunodeficiency syndromes, and congenital metabolic disorders. Nowadays, HSC transplantation is practiced in both allogeneic and autologous donations [66].

As mentioned before, the most important clinical challenge in HSCT is the lack of compatible donors in the golden time of transplant and also immunological rejection risk [67]. Subcutaneous injection of granulocyte colony stimulating factors (G-CSF), such as Filgrastim and Lenograstim, which are suspected to increase the risk of neoplasia, discharge HSC from BM to peripheral blood [68]. After Yamanaka studies in the field of reprogramming, which led to the invention iPS technology, many researchers have been trying to produce induced hematopoietic stem cells (iHSC) derived from iPSC [69]. In fact, iPS technology advancements have made the creation of healthy pluripotent (Subsequently multipotent) stem cells from patients with blood malignancy possible [70]. To produce these cells, Szabo et al. showed that the sole expression of OCT4 of OKSM factors can generate HSC from fibroblasts [69]. Doulatov et al. also reprogrammed myeloid cell line to HSC in a similar project using 13 transcription factors [70]. In the treatment of congenital-based leukemia, genetic disorders must be rectified. To this end, new genetic engineering tools can be used, including CRISPR-Cas9, zinc-finger nucleases (ZFN), meganucleases, transcription activator-like effector nucleases (TALEN) and double-strand break (DSB) nucleases [71-74]. In gene regulatory network complex, two transcription factors (HOXA and ERG) act as the main differentiation and proliferation factors in HSC. These two factors are inducers of the self-renewal ability and are thus oncogenic. Certainly, dysregulation of these factors develops a wide range of leukemic cells. Studies show that MYB, SOX4, RORA, HOXA5, and HOXA9 are all part of gene regulatory complex [70].

The classic marker of human iHSC is CD34 [75]. Many cluster of differentiation markers such as CD90, CD38, CD34, CD45, CD133, CD105, and C-kit (stem cell receptor) help distinguish HSC [76]. To ensure credible purification, cytochemistry and flow cytometry assays can be used, and the FACS technique is also useful to purify iPSCs and iHSCs. The safe, effective, and highly pure product ensures an HSCT with the least possibility of side effect.

6

155156157158

159160161162163164165166167

168169170171

172

173

174175176177178179180

181182183184185186187188189190191192193194195196

197198199200201

Page 7: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

3.2. Induced cytotoxic T-lymphocytes

During recent years, cancer immunotherapy has expanded by utilizing various technologies generally based on reversing immunosuppression [77-79], transferring cytotoxic T lymphocyte (CTL) [80-82], transferring TCR gene [83-85], and so on. Before producing iPSC-derived CTL (iCTL), it is possible to differentiate them into double positive cells (DP: CD4/8+) in op9 culture media [86, 87]. The major cytokine in T-cell differentiation is IL-2 [88, 89]. After DP purification, cells can be stimulated and differentiated to CD8αβ+. By analyzing RNA-seq, researchers realized that iCTL also expresses other surface markers including TBX21, RUNX3, GATA3, and IKZF1 in levels as high as CD8+ [87].

Due to the unlimited in-vitro expansion of iCTL, it is possible to provide iCTL based on the need of each therapeutic panel, and iPS technology has made it feasible to prepare autologous CTL from patients’ own somatic cells. When T-cell is reprogrammed from iPSC, the TCR gene, which responds to a specific antigen, is propagated inside the cell. Therefore, all iCTLs express the same TCR on their surface [90]. The iCTL has cytotoxic capabilities against particular proteins in leukemia, like Wilms’ Tumor protein1 (WT1) (in AML-HL60 and THP1 cell line). Certainly, cells that do not express MHC-I on their surface escape CTL cytotoxic effects even if WT1 is expressed. For the first time, iCTL was used to treat NOG mice with AML-HL60 cell line. The cured mice clearly showed longer lifespan compared with the control group. In the treated mice, iCTL was not oncogenic nor was it tumorigenic. Also, it did not initiate any tissue damage [87].

The advent of iPS technology and then the invention of iCTL have been an accomplishment to help us with new methods in the cellular immunotherapy of cancer. In addition to using iCTL against malignant cells in cancer and leukemia, it is also used in resistant infections. Considering high specificity of iCTL, lower risk of immunologic rejection (due to being autologous), and high efficiency and purity, it could develop a great shift in the treatment of cancer. iCTLs can be used as an agent to prevent cancer relapse. So, iCTL application is promising in treating and monitoring leukemic patients.

3.3. Reprogrammed NK cells

Natural killer cells (NK cells) are a part of immune system that secrete cytokines and enzymes to perforate and lyse invading cells. NK cells are the link between adaptive and innate immune systems that can enhance and accelerate immunologic responses using their memory ability [91].

Cell therapy has helped to improve the natural immune system by using ex-vivo cultured NK cells. Currently, these cells are extracted from the blood circulation by leukapheresis and then separated from other cells via magnetic beads coated with anti-CD56. After purification, it is possible to expand them in culture media with B cell line transformed with EBV, or K562 cell lines that express co-stimulatory molecules and IL-21 and IL-5 [92]. However, utilizing this ex-vivo growth method cannot produce stable NK cells in-vivo to confront malignant cells, so achieving a durable and efficient NK cell in-vitro is sorely needed [91]. According to the studies on induced NK cells based on iPSC (iNK cells), it is possible to acquire stable and effective NK cell in-vitro using IL-7 and FLT3L for differentiation [44, 49, 93]. For the first time, the use of iNK cells in-vivo has been successful in OVA mice (as a tumor of artificial Ag) and NOG mice (with AML-OVN1 and MEGO1 cell line) [91]. As in iCTL, reprogrammed NK cell can supply the desired volume of cell product to be used in leukemia therapy panel without any immunological response.

3.4. miRNA and iPSC

The microRNAs (miRNA) are a group of small interference RNA (19 to 22 nucleotide) that innately control protein-coding genes. Studies regarding this RNA has proven their role in the onset of tumorigenesis and tumor metastasis. Considering their role as oncogene and tumor suppressor, there are many possible therapeutic strategies to cure malignancies based on miRNA [94, 95].

7

202

203

204205206207208209210

211212213214215216217218219

220221222223224225

226

227

228229230

231232233234235236237238239240241

242

243244245246

Page 8: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

Induction of miRNAs correlated with oncogenic genes and inhibition of miRNAs related to tumor suppressor RNAs bursts progenitor cells count and starts malignancy. While miRNA is critical in intracellular processing, recent studies have emphasized its extracellular effects. These miRNAs are produced and secreted like hormones from cells in the microenvironment and are eventually released to blood circulation [96]. Secreted miRNA is taken up by cells in various parts of the body as they regulate protein expression prior to translation [97].

The iPS technology delivers new methods in miRNA-based therapies. As repeatedly said, iPS cells are pluripotent with self-renewal and differentiation potential. So, being able to derive them from hematopoietic cells and their ability to secrete particular miRNAs could pave the road in the future for target therapy of leukemia and other cancers [98]. Furthermore, miRNA has a direct role in regulating immune responses. For instance, transferring miR335 from T-cell to synaptic space created through Ag presentation has been done to neutralize intracellular reaction of target cell [99]. The iCTL (explained previously) with high expression potential of miR335, could be a part of compound therapy against malignant cells based on target therapy, immunotherapy, and cell therapy. In addition to the measures above, using miRNA has made it possible to combine iPS technology with current strategies in dealing with cancers, including sandwich RNAi inhibition, micro mimic agent complex RNAi inhibition, small molecules inhibitors of miRNAs (SMIRs), and targeting miRNA from microvesicle and exosome [94, 100].

4. Hematopoietic Malignancy Modeling

As mentioned before, one aspect of iPS technology is identifying the molecular pathway of different diseases to better understand their pathogenesis, and present distinctive therapy for them. Patients’ iPS cells, which differentiate to a specific cellular line of the current disease, can be used for in-vitro modeling, drug synthesis, appropriate dosage determination, and toxicity evaluation [101-103]. Separating the mutant cells from the bloodstream of the patient and reprogramming them to iPSC makes hematopoietic malignancy modeling possible. Being able to identify the translocation patterns in leukemic cells using only one cell derived from cancer progenitor cells is accomplished because of the stability of genetic content through reprogramming.

Currently, with access to these cells, assessing the epigenetics of malignancies and target therapy is within reach. For example, oncogenic translocations (including silencing of tumor suppressor genes, changing proto-oncogene to oncogene or dysregulation of apoptotic genes) and epigenetic dysregulations (changing DNA methylation pattern and modification of miRNA expression) can change the expression of progenitor cell genome leading to malignancy development. It has been shown that blocking the Hedgehog signaling could stop the growth of tumor in MM, CML, pancreas cancer, and brain tumors [104, 105]. Also, it has been demonstrated that NOTCH and Wnt signaling are involved in T-ALL, CML, and medulloblastoma [106]. In another example, human cancers generally show an increment of telomerase activity; this cellular process indicates that limiting telomerase activity likely prompts apoptosis and reduces self-renewal activity of mutant cells in MM [107].

So far, extensive leukemia modeling activities have been carried out using iPS technology including:

4.1. iPSC in AML: In 2017, Zhu et al. made the iPSC line derived from AML-M6 cells in-vitro and identified their biological properties and illustrated that these iPSCs are pluripotent and can differentiate into the three germ layers [108]. Also in 2015, Salci et al. produced HSC from fibroblasts of AML patients using iPS technology and compared their cytogenetic, immunophenotype, function, and morphology with their myeloid blasts. They showed that these HSCs have normal progenitor status and do not have leukemic mutations [109]. But In 2017, while Chao et al. were generating iPSC from MLL-rearranged AML patients, cells retained their cytogenetic and mutagenic abnormalities, but their epigenetic status had reversed back to normal. Therefore, there is no leukemic potential, but they regain the methylation patterns and leukemic properties upon re-differentiation to hematopoietic cells [110, 111]. In another study in 2014, Liu et al. succeed in making murine AML models. They created mice with chimeric iPSCs by overexpressing human MLL-AF9 fusion gene in hematopoietic cells containing OKSM factors. These mice spontaneously developed AML because of MLL-AF9 reactivation. This experiment showed that epigenetic

8

247248249250251

252253254255256257258259260261262

263

264

265266267268269270271

272273274275276277278279280

281

282283284285286287288289290291292

Page 9: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

regulators that manipulate the MLL-AF9 gene expression pattern are the bridge between MLL-AF9 leukemia cells and derived iPSCs [112].

4.2. iPSC in FPD/AML: In 2014, Sakurai et al. derived iPSCs from FPD/AML patients. They showed that in this situation hematopoietic progenitor cells’ formation and megakaryocyte differentiation are incomplete. For the first time, they showed the role of RUNX1 in human hematopoiesis and the effect of heterozygous mutant RUNX1 in megakaryocyte differentiation. They also showed that FPD/iPSC phenotypes are the result of the haploinsufficiency of RUNX1 [113]. In 2015, Lizuka et al. showed that RUNX1 is associated with thrombocytopenia in FPD/AML patients. Because HSC-derived FPD/iPSC had a dysregulation in megakaryocyte differentiation, and they corrected megakaryopoiesis by editing the RUNX1 mutation via TALEN [114].

4.3. iPSC in CML: In 2012, Kumano et al. produced iPSCs from imatinib-sensitive CML patients. These CML IPSCs continued to express the BCR-ABL onco-protein but were resistant to imatinib. They were shown to maintain iPSC signaling that compensate for the inhibition of BCR-ABL. Of course, these cells regained susceptibility to imatinib after being converted to the hematopoietic lineage [115].

4.4. iPSC in ALL: In 2016, Munoz-Lopez et al. attempted to produce iPSCs from the B-ALL subtypes, but neither the primary blasts nor the B-ALL cell lines (even with adding boosters) did not have reprogramming potential to achieve the pluripotency status. However, they were able to reprogram MLL-AF4-overexpressing HSCs/B-progenitors. This suggests that cells origin and leukemic genes were not the issue [116]. In 2016, Zhang et al. showed that the apoptosis, NF-κB, DOT1L, and LSD1 pathways were richer in reprogramming-incompetent colonies than reprogramming-competent colonies. The inhibitors of these pathways increased reprogramming efficiency in T-ALL cells. This indicates that these pathways are barriers to reprogramming in T-ALL cells [117].

4.5. iPSC in NS/JMML: In 2015, Mulero-Navarro et al. found that the myeloid cells derived from NS/JMML human induced pluripotent stem cells (hiPSC) had increased STAT5 signaling and two up-regulated microRNAs (miR-223 and miR-15a) that distinguish JMML caused by PTPN11 mutation from other genetic forms of this disease. Reducing the miR-223 activity causes normalization of the myelogenesis in the NS/JMML hiPSCs. Also, microRNA target gene expression levels were reduced in hiPSC-derived myeloid cells as well as in JMML cells with PTPN11 mutations (Table 2) [118].

Disease Year Type Origin Reprogramming factors

Vector Investigate Scientists References

CML 2012 Modeling - OKSM Retroviral CML modeling and study on pathophysiologic effects

Kumano K. et al. [115]

FPD/AML 2014 Modeling Peripheral T-cell

OKSM Sendaiviral Study on physiologic roles of RUNX1 in human hematopoiesis and FPD/AML pathophysiology

Sakurai M. et al. [113]

AML 2014 Modeling LIN- bone marrow cells

OKSM Retroviral Generation of AML-murine with overexpression of human MLL-AF9 fusion gene

Lui Y. et al. [112]

AML 2015 Modeling FibroblastOKSM

Lentiviral AML modeling and generating the healthy hematopoietic progenitors

Salci K.R. et al. [109]

NS/JMML 2015 Modeling Fibroblast OKSM Retroviral JMML modeling by human iPSC derived from NS/JMML and non-leukemic patients with PTPN11 mutation

Mulero-Navarro S. et al.

[118]

FPD/AML 2015 Modeling (gene editing)

Fibroblast OKSM Retroviral iPSC generation from a FPD/AML patient and modeling (with and without gene correction) via TALEN

Iizuka H et al. [114]

T-ALL 2016 Modeling LIN- bone marrow cells

OKSM Retroviral Identification of reprogramming blockage pathways in T-ALL

Zhang H. et al. [117]

9

293294

295296297298299300301

302303304305

306307308309310311312

313314315316317318

Page 10: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

B-ALL 2016 Modeling Myeloid cells

Polycistronics:1)lentiviral OKSM2)sendai OKSM

Monocystronics:1)Episomal OKSM or OKSL2)retroviral OKSM3)sendai OKSM

iPSC generation from B-ALL subtypes Munoz-Lopez A. et al.

[116]

AML(M6) 2017 Modeling Infiltrated skin

OKSM Lentiviral iPSC generation from AML cell line Zhu L.F. et al. [108]

AML 2017 Modeling-

OKSM Sendaiviral AML-iPSC generation and study on its epigenetic and genetic properties

Chao M.P. et al. [110]

Table 2. Recent iPS projects in the field of modeling of leukemia. NS: noonan syndrome; JMML: juvenile myelomonocytic leukemia; FPD/AML: familial platelet disorder with propensity to acute myeloid leukemia; LIN-: Lineage negative; OKSM: OCT3/4, SOX2, KLF4, c-MYC; OKSL: OCT3/4, SOX2, KLF4, LIN28.

Thus, understanding the pathogenesis and complete etiology of leukemia can help design more specialized drugs against malignant cells with lower side effects. For example, the drugs interfering with cell signaling will stimulate tumor suppressor genes or repress particular oncogenes. However, the systemic cytotoxicity of cell signal-interfering drugs has always been a major concern. It means that drug therapy influences both cancer cells and normal hematopoietic stem cells [107, 119, 120]. Providing malignant leukemia cells in a non-invasive way for in-vitro studies with great efficiency and in desirable abundance cannot be done so easily. This problem could be overcome by the application of iPS technology and reprogramming. Using malignant hematopoietic stem cells derived from iPSC will serve as a great tool in fighting leukemia in the fields of drug screening and monitoring, new drug designing, dosage determination for patients, and molecular studies.

5. Supportive Care by iPSC-derived Megakaryocytic and Erythropoietic Stem Cells

In most cases of leukemia, patients suffer from comorbidities resulting from the myelophthisic status. Occupying the existing niches in BM scaffolds by mutant and malignant cells diminishes the space needed for growth of normal cell types, such as erythroid cells and platelets. In addition, in some leukemias, such as AML-M6 and AML-M7, the direct destruction of erythroid and platelet lineage, respectively, are observed. Thus, in leukemic patients, symptoms related to erythropenia and thrombocytopenia, such as gastrointestinal hemorrhages, petechiae, purpura (and other coagulation disorders), respiratory distress (due to decreased RBC), lethargy, severe anemia, and drowsiness are frequently observed [122]. Sometimes, these symptoms are the main reason for patient referral to a physician, which is followed by leukemia diagnosis.

This problem could be overcome, in the future, by applying iPS technology. In 2012, Kobari et al. illustrated complete in-vitro terminal maturation of erythroid lineage from hiPSCs and subsequent normal production of Hb in-vivo by injection of hiPSCs into mice. This finding indicates that iPSC can be a supplemental source of RBCs. Their other goal was to produce modified functional RBCs from the sickle-cell IPSC without any gene editing and drug therapy [123]. In 2011, Dias et al. showed that the production of RBC from transgene-free human iPSC has similar efficiency compared with human embryonic stem cell (hESC). The Hb expression profiles were also similar to hESC-derived RBCs; however, the β-globin expression was lower than the embryonic and fetal globins after fibroblasts transition to pluripotency status [124]. In 2016, Borger et al. showed that iPSCs that their HLA class I expression was silenced can effectively produce megakaryocyte (MK) and functional platelets with low immunogenicity. This is important because the most common cause of immunological platelet transfusion refractoriness is HLA-I-sensitive alloantibodies. Due to their ability to survive antibody-mediated complement and cell-dependent cytotoxicity, they can be used in thrombocytopenic patients with or without alloimmunity [125]. In 2014, Feng et al. described a rapid protocol for differentiation of hiPSC into megakaryocytes and functional platelets, resulting in the production of large amounts of cryopreservable MK progenitors. Hence, it gives the advantage of having countless platelets at a moment's notice upon thawing these cells [126].

10

319

320321322

323324325326327328329330331

332

333

334335336337338339340341

342343344345346347348349350351352353354355356

Page 11: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

The unlimited proliferation of iPSC in-vitro not only provides desirable amounts of megakaryocyte and erythrocyte progenitors, but it could also generate other healthy cell lines, such as WBC. This could be used in lymphopenia or myelophthisic status in which the cell count and immune response are diminished. After receiving mature somatic cells from patients and reprogramming and differentiating them to HSC, first, the extracted HSCs could be differentiated to myeloid progenitors by IL-3, IL-1, GM-CSF, SCF, and IL-6. Then, some progenitors could be given megakaryocyte-stimulating cytokines like IL-3, hG-CSF, TPO, and SCF; and some could be given erythrocyte-stimulating cytokines like EPO, IL-3, hGM-CSF, and SCF depending on the purpose (Figure 1) [127].

Figure 1. iPS technology application in leukemia. First, somatic cells are reprogrammed to iPSC. The iHSCs are the first differentiated cells. Different cytokines drive differentiation to various immunologic cells like RBC, platelet, and malignant simulated cells

The autologous transplantation can be applied in non-severe cases using the aforementioned progenitors or hematopoietic products (RBC and PLT) without immunological rejection risk or other transmitted diseases. In urgent situations, pre-prepared heterologous products of iPSCs with the lowest expression of surface antigens can be used. Controllable expression of surface antigens in the iPSC-derived RBCs and PLTs enables a safe, heterologous transfusion. Evidently, using iPS technology in this field makes it possible to overcome many erythrocytopathies and thrombocytopathies in addition to leukemia.

Conclusion and Future Perspective

11

357358359360361362363

364

365366367

368369370371372373

374

375

Page 12: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

As mentioned before, iPSC serving as a new invention in clinical field is an effective advancement in therapy and investigating the background of various diseases. Regarding new ideas and prospective projects, we have only scratched the surface of what we can do with it. In the future, advancement in research projects and suggestion of new theories will be observed in the treatment of many maladies like leukemia. The financial costs and mortalities will be reduced after routinely executing iPS technology in therapeutic clinics. Considered ideas and projects in this paper are in this direction. Besides leukemia, the mentioned iCTLs and iNK cells could be used extensively in the field of treatment by providing specificity against a particular pathogen. The invention of iPSC has also demonstrated promising results in different dimensions of the molecular biology of miRNA. The future world of iPSC will be significant and profound, and the power of imagination will propel this technology.

Conflict of Interest

The authors declare that there are no conflicts of interest.

Acknowledgement

We would like to thank Dr. Atashi and Dr. Azad for their valuable input on this article.

References

[1] Azad M, Bakhshi Biniaz R, Goudarzi M, et al. Short view of leukemia diagnosis and treatment in iran. Int J Hematol Oncol Stem Cell Res 2015; 9(2): 88-94.

[2] Meyskens FL, Jr., Mukhtar H, Rock CL, et al. Cancer Prevention: Obstacles, Challenges and the Road Ahead. J Natl Cancer Inst 2016; 108(2).

[3] Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol 2013; 15(4): 338-44.

[4] Alexis F, Rhee JW, Richie JP, Radovic-Moreno AF, Langer R, Farokhzad OC. New frontiers in nanotechnology for cancer treatment. Urol Oncol 2008; 26(1): 74-85.

[5] Seigneuric R, Markey L, Nuyten DS, et al. From nanotechnology to nanomedicine: applications to cancer research. Curr Mol Med 2010; 10(7): 640-52.

[6] Pakakasama S, Kajanachumpol S, Kanjanapongkul S, et al. Simple multiplex RT-PCR for identifying common fusion transcripts in childhood acute leukemia. Int J Lab Hematol 2008; 30(4): 286-91.

12

376377378379380381382383384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400401

402403

404

405406

407408

409410411

Page 13: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[7] Stams WA, den Boer ML, Beverloo HB, et al. Expression levels of TEL, AML1, and the fusion products TEL-AML1 and AML1-TEL versus drug sensitivity and clinical outcome in t(12;21)-positive pediatric acute lymphoblastic leukemia. Clin Cancer Res. 2005; 11(8): 2974-80.

[8] McWhirter JR, Neuteboom ST, Wancewicz EV, Monia BP, Downing JR, Murre C. Oncogenic homeodomain transcription factor E2A-Pbx1 activates a novel WNT gene in pre-B acute lymphoblastoid leukemia. Proc Natl Acad Sci U S A 1999; 96(20): 11464-9.

[9] Rudolph C, Hegazy AN, von Neuhoff N, et al. Cytogenetic characterization of a BCR-ABL transduced mouse cell line. Cancer Genet Cytogenet 2005; 161(1): 51-6.

[10] Caslini C, Serna A, Rossi V, Introna M, Biondi A. Modulation of cell cycle by graded expression of MLL-AF4 fusion oncoprotein. Leukemia 2004; 18(6): 1064-71.

[11] Martin-Subero JI, Odero MD, Hernandez R, et al. Amplification of IGH/MYC fusion in clinically aggressive IGH/BCL2-positive germinal center B-cell lymphomas. Genes Chromosomes Cancer. 2005; 43(4): 414-23.

[12] Rafiei A, Mian AA, Doring C, et al. The functional interplay between the t(9;22)-associated fusion proteins BCR/ABL and ABL/BCR in Philadelphia chromosome-positive acute lymphatic leukemia. PLoS Genet 2015; 11(4): e1005144.

[13] Zalcberg IQ, Silva ML, Abdelhay E, et al. Translocation 11;14 in three children with acute lymphoblastic leukemia of T-cell origin. Cancer Genet Cytogenet. 1995; 84(1): 32-8.

[14] Rahmani T, Azad M, Chahardouli B, Nasiri H, Vatanmakanian M, Kaviani S. Patterns of DNMT1 Promoter Methylation in Patients with Acute Lymphoblastic Leukemia. Int J Hematol Oncol Stem Cell Res 2017; 11(3): 172-7.

[15] Chiaretti S, Zini G, Bassan R. Diagnosis and subclassification of acute lymphoblastic leukemia. Mediterr J Hematol Infect Dis 2014; 6(1): e2014073.

[16] McGregor S, McNeer J, Gurbuxani S. Beyond the 2008 World Health Organization classification: the role of the hematopathology laboratory in the diagnosis and management of acute lymphoblastic leukemia. Semin Diagn Pathol 2012; 29(1): 2-11.

[17] Porwit A, Bene MC. Acute leukemias of ambiguous origin. Am J Clin Pathol 2015; 144(3): 361-76.

[18] Faderl S, O'Brien S, Pui CH, et al. Adult acute lymphoblastic leukemia: concepts and strategies. Cancer 2010; 116(5): 1165-76.

[19] Bassan R, Hoelzer D. Modern therapy of acute lymphoblastic leukemia. J Clin Oncol. 2011; 29(5): 532-43.

[20] Hladnik L, Augustin K, DeFrates S. Advancements in Therapy for Acute Lymphoblastic Leukemia: Blinatumomab J Adv Pract Oncol 2016; 7(1): 76-82.

[21] Zenz T, Vollmer D, Trbusek M, et al. TP53 mutation profile in chronic lymphocytic leukemia: evidence for a disease specific profile from a comprehensive analysis of 268 mutations Leukemia 2010; 24(12): 2072-9.

13

412413414

415416417

418419

420421

422423424

425426427

428429

430431432

433434

435436437

438439

440441

442443

444445

446447448

Page 14: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[22] Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2002; 99(24): 15524-9.

[23] Chiorazzi N, Rai KR, Ferrarini M. Chronic lymphocytic leukemia. N Engl J Med. 2005; 352(8): 804-15.

[24] Marton S, Garcia MR, Robello C, et al. Small RNAs analysis in CLL reveals a deregulation of miRNA expression and novel miRNA candidates of putative relevance in CLL pathogenesis. Leukemia 2008; 22(2): 330-8.

[25] Barcena C, Quesada V, De Sandre-Giovannoli A, et al. Exome sequencing identifies a novel mutation in PIK3R1 as the cause of SHORT syndrome. BMC Med Genet 2014; 15:51.

[26] Zenz T, Mertens D, Kuppers R, Dohner H, Stilgenbauer S. From pathogenesis to treatment of chronic lymphocytic leukaemia. Nat Rev Cancer 2010; 10(1): 37-50.

[27] Seiffert M, Dietrich S, Jethwa A, Glimm H, Lichter P, Zenz T. Exploiting biological diversity and genomic aberrations in chronic lymphocytic leukemia. Leuk Lymphoma 2012; 53(6): 1023-31.

[28] Hallek M, Fischer K, Fingerle-Rowson G, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 2010; 376(9747): 1164-74.

[29] Melo JV, Catovsky D, Gregory WM, Galton DA. The relationship between chronic lymphocytic leukaemia and prolymphocytic leukaemia. IV. Analysis of survival and prognostic features. Br J Haematol 1987; 65(1): 23-9.

[30] Challen GA, Boles NC, Chambers SM, Goodell MA. Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-beta1. Cell Stem Cell 2010; 6(3): 265-78.

[31] Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med 2017; 377(5): 454-64.

[32] Meyer SC, Levine RL. Translational implications of somatic genomics in acute myeloid leukaemia. Lancet Oncol 2014; 15(9): e382-94.

[33] Medinger M, Lengerke C, Passweg J. Novel therapeutic options in Acute Myeloid Leukemia. Leuk Res Rep 2016; 6: 39-49.

[34] Medinger M, Lengerke C, Passweg J. Novel Prognostic and Therapeutic Mutations in Acute Myeloid Leukemia. Cancer Genomics Proteomics 2016; 13(5): 317-29.

[35] Bronkhorst AJ, Wentzel JF, Aucamp J, van Dyk E, du Plessis L, Pretorius PJ. Characterization of the cell-free DNA released by cultured cancer cells. Biochim Biophys Acta 2016; 1863(1): 157-65.

[36] Shibata N, Miyamoto N, Nagai K, et al. Development of protein degradation inducers of oncogenic BCR-ABL protein by conjugation of ABL kinase inhibitors and IAP ligands. Cancer Sci. 2017; 108(8): 1657-66.

[37] Zomer HD, Vidane AS, Goncalves NN, Ambrosio CE. Mesenchymal and induced pluripotent stem cells: general insights and clinical perspectives. Stem Cells Cloning 2015; 8: 125-34.

14

449450451

452453

454455456

457458

459460

461462

463464465

466467468

469470

471472

473474

475476

477478

479480

481482483

484485

Page 15: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[38] Mitalipov S, Wolf D. Totipotency, pluripotency and nuclear reprogramming. Adv Biochem Eng Biotechnol 2009; 114: 185-99.

[39] Nadri S, Soleimani M, Hosseni RH, Massumi M, Atashi A, Izadpanah R. An efficient method for isolation of murine bone marrow mesenchymal stem cells. Int J Dev Biol 2007; 51(8): 723-9.

[40] Nichols J, Zevnik B, Anastassiadis K, et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 1998; 95(3): 379-91.

[41] Mitsui K, Tokuzawa Y, Itoh H, et al. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 2003; 113(5): 631-42.

[42] Rad SM, Mohammadi-Sangcheshmeh A, Bamdad T, et al. Pluripotency Crossroads: Junction of Transcription Factors, Epigenetic Mechanisms, MicroRNAs, and Long Non-coding RNAs. Curr Stem Cell Res Ther 2017; 12(4): 300-11.

[43] Seah YF, El Farran CA, Warrier T, Xu J, Loh YH. Induced Pluripotency and Gene Editing in Disease Modelling: Perspectives and Challenges. Int J Mol Sci 2015; 16(12): 28614-34.

[44] Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126(4): 663-76.

[45] Duinsbergen D, Salvatori D, Eriksson M, Mikkers H. Tumors originating from induced pluripotent stem cells and methods for their prevention. Ann N Y Acad Sci 2009; 1176: 197-204.

[46] Lin T, Ambasudhan R, Yuan X, et al. A chemical platform for improved induction of human iPSCs. Nat Methods 2009; 6(11): 805-8.

[47] Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007; 318(5858): 1917-20.

[48] Maherali N, Ahfeldt T, Rigamonti A, Utikal J, Cowan C, Hochedlinger K. A high-efficiency system for the generation and study of human induced pluripotent stem cells. Cell Stem Cell 2008; 3(3): 340-5.

[49] Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature 2007; 448(7151): 313-7.

[50] Merkl C, Saalfrank A, Riesen N, et al. Efficient generation of rat induced pluripotent stem cells using a non-viral inducible vector. PLoS One 2013; 8(1): e55170.

[51] Hou P, Li Y, Zhang X, et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 2013; 341(6146): 651-4.

[52] Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc Jpn Acad Ser B Phys Biol Sci 2009; 85(8): 348-62.

[53] Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 2010; 7(5): 618-30.

15

486487

488489

490491

492493

494495496

497498

499500

501502

503504

505506

507508509

510511

512513

514515

516517518

519520521

Page 16: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[54] Chang CW, Lai YS, Pawlik KM, et al. Polycistronic lentiviral vector for "hit and run" reprogramming of adult skin fibroblasts to induced pluripotent stem cells. Stem Cells 2009; 27(5): 1042-9.

[55] Yusa K, Rad R, Takeda J, Bradley A. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon. Nat Methods 2009; 6(5): 363-9.

[56] Zhou H, Wu S, Joo JY, et al. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell. 2009; 4(5): 381-4.

[57] Kim D, Kim CH, Moon JI, et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 2009; 4(6): 472-6.

[58] Lee CH, Kim JH, Lee HJ, et al. The generation of iPS cells using non-viral magnetic nanoparticle based transfection. Biomaterials 2011; 32(28): 6683-91.

[59] Phillips MD, Kuznetsov SA, Cherman N, et al. Directed differentiation of human induced pluripotent stem cells toward bone and cartilage: in vitro versus in vivo assays. Stem Cells Transl Med 2014; 3(7): 867-78.

[60] Bilousova G, Jun du H, King KB, et al. Osteoblasts derived from induced pluripotent stem cells form calcified structures in scaffolds both in vitro and in vivo. Stem Cells 2011; 29(2): 206-16.

[61] Mari C, Winyard P. Concise Review: Understanding the Renal Progenitor Cell Niche In Vivo to Recapitulate Nephrogenesis In Vitro. Stem Cells Transl Med 2015; 4(12): 1463-71.

[62] Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 2007; 318(5858): 1920-3.

[63] Birbrair A, Frenette PS. Niche heterogeneity in the bone marrow. Ann N Y Acad Sci 2016; 1370(1): 82-96.

[64] Cutler C, Antin JH. Peripheral blood stem cells for allogeneic transplantation: a review. Stem Cells 2001; 19(2): 108-17.

[65] Langroudi L, Forouzandeh M, Soleimani M, Atashi A, Golestaneh AF. Induction of differentiation by down-regulation of Nanog and Rex-1 in cord blood derived unrestricted somatic stem cells. Mol Biol Rep 2013; 40(7): 4429-37.

[66] Bruno B, Rotta M, Patriarca F, et al. A comparison of allografting with autografting for newly diagnosed myeloma. N Engl J Med 2007; 356(11): 1110-20.

[67] Sundaramurthi T, Wehrlen L, Friedman E, Thomas S, Bevans M. Hematopoietic Stem Cell Transplantation Recipient and Caregiver Factors Affecting Length of Stay and Readmission. Oncol Nurs Forum 2017; 44(5): 571-9.

[68] Sorror ML, Maris MB, Storb R, et al. Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT. Blood 2005; 106(8): 2912-9.

[69] Szabo E, Rampalli S, Risueno RM, et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature 2010; 468(7323): 521-6.

16

522523524

525526

527528

529530

531532

533534535

536537

538539

540541

542543

544545

546547548

549550

551552553

554555

556557

Page 17: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[70] Doulatov S, Vo LT, Chou SS, et al. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell 2013; 13(4): 459-70.

[71] Perez-Pinera P, Ousterout DG, Gersbach CA. Advances in targeted genome editing. Curr Opin Chem Biol 2012; 16(3-4): 268-77.

[72] Kaufmann KB, Buning H, Galy A, Schambach A, Grez M. Gene therapy on the move. EMBO Mol Med 2013; 5(11): 1642-61.

[73] Munoz IG, Prieto J, Subramanian S, et al. Molecular basis of engineered meganuclease targeting of the endogenous human RAG1 locus. Nucleic Acids Res 2011; 39(2): 729-43.

[74] Hotta A, Yamanaka S. From Genomics to Gene Therapy: Induced Pluripotent Stem Cells Meet Genome Editing. Annu Rev Genet 2015; 49: 47-70.

[75] Civin CI, Strauss LC, Brovall C, Fackler MJ, Schwartz JF, Shaper JH. Antigenic analysis of hematopoiesis. III. A hematopoietic progenitor cell surface antigen defined by a monoclonal antibody raised against KG-1a cells. J Immunol 1984; 133(1): 157-65.

[76] Lo YM, Corbetta N, Chamberlain PF, et al. Presence of fetal DNA in maternal plasma and serum. Lancet 1997; 350(9076): 485-7.

[77] Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363(8): 711-23.

[78] Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366(26): 2443-54.

[79] Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369(2): 122-33.

[80] Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011; 17(13): 4550-7.

[81] Chapuis AG, Ragnarsson GB, Nguyen HN, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med 2013; 5(174): 174ra27.

[82] Doubrovina E, Oflaz-Sozmen B, Prockop SE, et al. Adoptive immunotherapy with unselected or EBV-specific T cells for biopsy-proven EBV+ lymphomas after allogeneic hematopoietic cell transplantation. Blood 2012; 119(11): 2644-56.

[83] Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006; 314(5796): 126-9.

[84] Johnson LA, Morgan RA, Dudley ME, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009; 114(3): 535-46.

17

558559560

561562

563564

565566

567568

569570571

572573

574575

576577

578579

580581582

583584585

586587588

589590

591592593

Page 18: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[85] Robbins PF, Morgan RA, Feldman SA, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 2011; 29(7): 917-24.

[86] Kovalovsky D, Uche OU, Eladad S, et al. The BTB-zinc finger transcriptional regulator PLZF controls the development of invariant natural killer T cell effector functions. Nat Immunol 2008; 9(9): 1055-64.

[87] Maeda T, Nagano S, Ichise H, et al. Regeneration of CD8alphabeta T Cells from T-cell-Derived iPSC Imparts Potent Tumor Antigen-Specific Cytotoxicity. Cancer Res 2016; 76(23): 6839-50.

[88] Hivroz C, Chemin K, Tourret M, Bohineust A. Crosstalk between T lymphocytes and dendritic cells. Crit Rev Immunol 2012; 32(2): 139-55.

[89] Hoyer S, Prommersberger S, Pfeiffer IA, et al. Concurrent interaction of DCs with CD4(+) and CD8(+) T cells improves secondary CTL expansion: It takes three to tango. Eur J Immunol 2014; 44(12): 3543-59.

[90] Vizcardo R, Masuda K, Yamada D, et al. Regeneration of human tumor antigen-specific T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell 2013; 12(1): 31-6.

[91] Fujii S, Shimizu K, Okamoto Y, et al. NKT cells as an ideal anti-tumor immunotherapeutic. Front Immunol 2013; 4: 409.

[92] Kreslavsky T, Savage AK, Hobbs R, et al. TCR-inducible PLZF transcription factor required for innate phenotype of a subset of gammadelta T cells with restricted TCR diversity. Proc Natl Acad Sci U S A 2009; 106(30): 12453-8.

[93] Watarai H, Fujii S, Yamada D, et al. Murine induced pluripotent stem cells can be derived from and differentiate into natural killer T cells. J Clin Invest 2010; 120(7): 2610-8.

[94] Shah MY, Ferrajoli A, Sood AK, Lopez-Berestein G, Calin GA. microRNA Therapeutics in Cancer - An Emerging Concept. EBioMedicine 2016; 12: 34-42.

[95] Golestaneh AF, Atashi A, Langroudi L, Shafiee A, Ghaemi N, Soleimani M. miRNAs expressed differently in cancer stem cells and cancer cells of human gastric cancer cell line MKN-45. Cell Biochem Funct. 2012; 30(5): 411-8.

[96] Shah MY, Calin GA. The mix of two worlds: non-coding RNAs and hormones. Nucleic Acid Ther 2013; 23(1): 2-8.

[97] Tominaga N, Kosaka N, Ono M, et al. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat Commun 2015; 6: 6716.

[98] Chen L, Mizutani A, Kasai T, et al. Mouse induced pluripotent stem cell microenvironment generates epithelial-mesenchymal transition in mouse Lewis lung cancer cells. Am J Cancer Res. 2014; 4(1): 80-8.

[99] Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun 2011; 2: 282.

18

594595596

597598599

600601

602603

604605606

607608

609610

611612613

614615

616617

618619620

621622

623624

625626627

628629

Page 19: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[100] Sahmani M, Vatanmakanian M, Goudarzi M, Mobarra N, Azad M. Microchips and their Significance in Isolation of Circulating Tumor Cells and Monitoring of Cancers. Asian Pac J Cancer Prev 2016; 17(3): 879-94.

[101] Arai S, Miyauchi M, Kurokawa M. Modeling of hematologic malignancies by iPS technology. Exp Hematol 2015; 43(8): 654-60.

[102] Hafizi M, Bakhshandeh B, Soleimani M, Atashi A. Exploring the enkephalinergic differentiation potential in adult stem cells for cell therapy and drug screening implications. In Vitro Cell Dev Biol Anim 2012; 48(9): 562-9.

[103] Houshmand M, Soleimani M, Atashi A, Saglio G, Abdollahi M, Nikougoftar Zarif M. Mimicking the Acute Myeloid Leukemia Niche for Molecular Study and Drug Screening. Tissue Eng Part C Methods 2017; 23(2): 72-85.

[104] Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr Biol 2007; 17(2): 165-72.

[105] Fouladiha H, Marashi SA, Shokrgozar MA, Farokhi M, Atashi A. Applications of a metabolic network model of mesenchymal stem cells for controlling cell proliferation and differentiation. Cytotechnology 2018; 70(1): 331-8.

[106] Lin T, Jones RJ, Matsui W. Cancer stem cells: relevance to SCT. Bone Marrow Transplant 2009; 43(7): 517-23.

[107] Zhihua Z, Miaomiao S, Jianping Y, Kuisheng C. Research for the influence of telomerase inhibitors on myeloma cell and therapy. Pak J Pharm Sci 2014; 27(5).

[108] Zhu LF, Chen QR, Chen SZ, et al. The Construction and Identification of Induced Pluripotent Stem Cells Derived from Acute Myelogenous Leukemia Cells. Cell Physiol Biochem 2017; 41(4): 1661-74.

[109] Salci KR, Lee JH, Laronde S, et al. Cellular Reprogramming Allows Generation of Autologous Hematopoietic Progenitors From AML Patients That Are Devoid of Patient-Specific Genomic Aberrations. Stem Cells 2015; 33(6): 1839-49.

[110] Chao MP, Gentles AJ, Chatterjee S, et al. Human AML-iPSCs Reacquire Leukemic Properties after Differentiation and Model Clonal Variation of Disease. Cell Stem Cell 2017; 20(3): 329-44 e7.

[111] Azad M, Kaviani S, Noruzinia M, et al. Gene Expression Status and Methylation Pattern in Promoter of P15INK4b and P16INK4a in Cord Blood CD34 (+) Stem Cells. Iran J Basic Med Sci 2013; 16(7): 822-8.

[112] Liu Y, Cheng H, Gao S, et al. Reprogramming of MLL-AF9 leukemia cells into pluripotent stem cells. Leukemia 2014; 28(5): 1071-80.

[113] Sakurai M, Kunimoto H, Watanabe N, et al. Impaired hematopoietic differentiation of RUNX1-mutated induced pluripotent stem cells derived from FPD/AML patients. Leukemia 2014; 28(12): 2344-54.

19

630631632

633634

635636637

638639640

641642643

644645646

647648

649650

651652653

654655656

657658

659660661

662663

664665666

Page 20: bentham.manuscriptpoint.com · Web viewLeukemia is an uncontrollable growth of hematopoietic cells due to a mutation in DNA followed by cellular dysregulation and one or more chromosomal

[114] Iizuka H, Kagoya Y, Kataoka K, et al. Targeted gene correction of RUNX1 in induced pluripotent stem cells derived from familial platelet disorder with propensity to myeloid malignancy restores normal megakaryopoiesis. Exp Hematol 2015; 43(10): 849-57.

[115] Kumano K, Arai S, Hosoi M, et al. Generation of induced pluripotent stem cells from primary chronic myelogenous leukemia patient samples. Blood 2012; 119(26): 6234-42.

[116] Munoz-Lopez A, Romero-Moya D, Prieto C, et al. Development Refractoriness of MLL-Rearranged Human B Cell Acute Leukemias to Reprogramming into Pluripotency. Stem Cell Reports 2016; 7(4): 602-18.

[117] Zhang H, Cheng H, Wang Y, et al. Reprogramming of Notch1-induced acute lymphoblastic leukemia cells into pluripotent stem cells in mice. Blood Cancer J 2016; 6(7): e444.

[118] Mulero-Navarro S, Sevilla A, Roman AC, et al. Myeloid Dysregulation in a Human Induced Pluripotent Stem Cell Model of PTPN11-Associated Juvenile Myelomonocytic Leukemia. Cell Rep 2015; 13(3): 504-15.

[119] Guzman ML, Rossi RM, Neelakantan S, et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 2007; 110(13): 4427-35.

[120] Guzman ML, Neering SJ, Upchurch D, et al. Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 2001; 98(8): 2301-7.

[121] Gibbins FJ. Haematological problems in the older patient. Practitioner 1975; 215(1289): 606-2.

[122] Kobari L, Yates F, Oudrhiri N, et al. Human induced pluripotent stem cells can reach complete terminal maturation: in vivo and in vitro evidence in the erythropoietic differentiation model. Haematologica 2012; 97(12): 1795-803.

[123] Dias J, Gumenyuk M, Kang H, et al. Generation of red blood cells from human induced pluripotent stem cells. Stem Cells Dev 2011; 20(9): 1639-47.

[124] Borger AK, Eicke D, Wolf C, et al. Generation of HLA-universal iPSCs-derived megakaryocytes and platelets for survival under refractoriness conditions. Mol Med 2016; 22: 274.

[125] Feng Q, Shabrani N, Thon JN, et al. Scalable generation of universal platelets from human induced pluripotent stem cells. Stem Cell Reports 2014; 3(5): 817-31.

[126] Easterbrook J, Fidanza A, Forrester LM. Concise review: programming human pluripotent stem cells into blood. Br J Haematol 2016; 173(5): 671-9.

20

667668669

670671

672673674

675676

677678679

680681682

683684

685

686687688

689690

691692

693694

695696

697