Anda di halaman 1dari 19

Hindawi Stem Cells International Volume

2018, Article ID 5416923, 16 halaman


https://doi.org/10.1155/2018/5416923Cell

Tinjau Artikel Stem Cancer (CSCs) dalam Resistensi


Obat dan Implikasi Terapi dalam Pengobatan Kanker

Lan Thi Hanh Phi, Ita Novita Sari, Ying-Gui Yang, Sang-Hyun Lee, Nayoung Jun, Kim
Kwang Seock, Yun Kyung Lee, dan Hyog Young Kwon

Soonchunhyang Institut Ilmu Medi-bio (SIMS), Universitas Soonchunhyang, Asan, Republik Korea.

Korespondensi harus ditujukan kepada Yun Kyung Lee; yunklee@sch.ac.kr dan Hyog Young Kwon; hykwon@sch.ac.kr

Menerima 28 September 2017; Diterima 11 Januari 2018; Diterbitkan 28


Februari 2018

Editor Akademik: Pratima Basak

Hak Cipta © 2018 Lan Thi Hanh Phi et al. Ini adalah artikel akses terbuka yang didistribusikan di bawah Lisensi Atribusi Creative
Commons, yang memungkinkan penggunaan, distribusi, dan reproduksi tanpa batas dalam media apa pun, asalkan karya aslinya
dikutip dengan benar.

Sel-sel kanker stem (CSC), juga dikenal sebagai sel-sel tumor-memulai (tics), disarankan untuk bertanggung jawab untuk resistensi
obat dan kekambuhan kanker karena sebagian untuk kemampuan mereka untuk memperbaharui diri sendiri dan differentiate ke
garis keturunan heterogen sel kanker . Dengan demikian, penting untuk memahami karakteristik dan mekanisme yang digunakan
CSC untuk menunjukkan resistensi terhadap agen terapi. Dalam ulasan ini, kami menyoroti fitur utama dan mekanisme yang
mengatur fungsi CSC dalam resistensi obat serta terobosan terbaru dari pendekatan terapeutik untuk menargetkan CSC. Ini
menjanjikan wawasan baru CSCs dalam resistensi obat dan memberikan alasan terapi yang lebih baik untuk menemani terapi
antikanker baru.
tubuh, sementara menggabungkan radioterapi dengan
kemoterapi dapat effectively memberikan hasil yang lebih baik
untuk mengobati berbagai jenis kanker [3]. Agen kemoterapi
1. Pendahuluan terbaru berhasil melawan lesi tumor primer dan residunya
setelah operasi atau radioterapi [4]. Namun, kemoterapi
Kanker adalah salah satu penyebab utama morbiditas dan menyebabkan heterogenitas tumor berasal dari kedua normal
mortalitas di seluruh dunia dengan sekitar 20% dari semua dan sel kanker dan heterogenitas dalam tumor, pada gilirannya,
kematian di negara maju [1]. Dari studi kanker praklinis dan menghasilkan mengurangi effEctskemoterapi; berkontribusi
klinis, berbagai pilihan diagnostik dan pengobatan baru untuk terhadap kegagalan pengobatan dan perkembangan penyakit
pasien kanker memberikan kemajuan penting dalam [5, 6]. Chemoresistance adalah masalah utama dalam
pengobatan dan pencegahan kanker [2]. Heterogenitas kanker pengobatan pasien kanker, sel-sel kanker menjadi resisten
adalah salah satu alasan yang berkontribusi terhadap kegagalan terhadapkimia
pengobatan dan perkembangan penyakit. Di antara beberapa zatyang digunakan dalam pengobatan, yang akibatnya
perawatan kanker, perawatan utama yang biasa digunakan membatasi eFFIefisiensi agen kemoterapi [7]. Ini juga sering
untuk merawat pasien adalah pembedahan, radioterapi, dan dikaitkan dengan tumor yang berubah menjadi bentuk yang
kemoterapi. Bedah berhasil dapat menghapus kanker dari lebih agresif dan / atau tipe metastasis [8-11].
Mengumpulkan bukti-bukti menunjukkan bahwa stem2. Penanda Asal dan PermukaanKanker
sel kanker (CSC) populasi, subkelompok sel kanker, Sel Punca(CSC)
bertanggung jawab untuk chemoresistance dan kanker
kambuh, karena memiliki kemampuan untuk memperbaharuiSel punca kanker (CSC), juga dikenal sebagai sel pemicu
diri dan differentiate ke dalam garis keturunan heterogen seltumor (TIC), telah dipelajari secara intensif dalam dekade
kanker dalam menanggapi agen kemoterapi [12-14]. CSC jugaterakhir, dengan fokus pada sumber yang mungkin, asal,
mampu menginduksi siklus sel tahanan (state diam) yangseluler penanda, studi mekanisme, dan pengembangan strategi
mendukung kemampuan mereka untuk menjadi resistenterapi yang menargetkan jalur mereka [23, 24]. Yangpertama
terhadap kemoterapi dan radioterapi[15-20].Agen-agenmeyakinkan-bukti dence dari CSC dilaporkan oleh Bonnet dan
kemoterapi umum menargetkan sel-sel yang berproliferasiDick pada tahun 1997 olehpengidentifikasiandari
untuk memimpin apoptosis mereka, seperti yang disebutkansubpopulasi sel leukemia mengekspresikan permukaan
sebelumnya. Meskipun terapi kanker yang berhasilpenanda CD34, tapi tidak CD38. CD34+/ CD38- subpopulasi
menghapuskan sebagian besar sel-sel tumor yang berkembangmampu memulai pertumbuhan tumor pada tikus penerima
biak, sejumlah CSC yang tersisa dapat bertahan danNOD / SCID setelah transplantasi [25]. Selain kanker darah,
meningkatkan kekambuhan kanker karena kemampuan merekaCSC telahdiidentifikasidalam beberapa jenis tumor padat [21,
untuk membangun invasifitas yang lebih tinggi dan26]. pertamaBukti dari kehadiran CSC pada kanker padat in
chemoresistance [21, 22]. Memahami fitur-fitur CSC penting
vivo ditemukan dandiidentifikasisebagai CD44+CD24-
untuk membangun fondasi bagi era baru dalam pengobatan/rendah
Silsilah- sel pada tikus berjanji immunocom- setelah
kanker. Dalam ulasan ini, kami membahas mekanisme rinci di
transplantasi sel kanker payudara manusia pada tahun 2003
mana CSC menampilkan resistensi terhadap kemoterapi dan
[27] meskipun memiliki telah ditunjukkan secara in vitro pada
radioterapi dan implikasinya untuk uji klinis.
tahun 2002 oleh penemuan sel-sel klonogenik (pembentuk
bola) yang diisolasi dari glioma otak manusia [28]. Seiring
waktu, populasi CSC jugadiidentifikasidari beberapa petugas
bisa- padat lainnya termasuk melanoma, otak, paru-paru, hati,
pankreas, usus besar, kanker payudara, serta kanker ovarium
[27, 29-35].
Meskipun model CSC menjelaskan heterogenitas
kanker dalam hal struktur hierarkis danperkembangan
mode, asal-usul CSC saat ini tidak jelas dan kontroversial [36,
37]. Mengumpulkan hipotesis menunjukkan bahwa tergantung
pada jenis tumor, CSC mungkin berasal dari salah satu sel
induk dewasa, sel-sel progenitor dewasa yang telah mengalami
mutasi, atau dari differentiated sel / sel cer bisa- yang diperoleh
sifat batang-seperti melalui dedif- ferentiation [ 25, 38-50].
Karena plastisitas CSC, telah menyarankan bahwa APY ther-
kombinasional menargetkan jalur CSC dan terapi kemo
konvensional mungkin memiliki lebih baik terapi effect, yang
akan dijelaskan nanti secara rinci (Gambar 1). Studi awal di
AML menunjukkan bahwa sel-sel primitif normal, tetapi tidak
terikat pada sel progenitor, adalah target untuk transformasi
leukemia [25]. Demikian pula, telah diindikasikan bahwa
penghapusan APC dalam Lgr5+ (ulangi kaya leusin yang
mengandung reseptor ditambah G-protein 5) sel-sel induk usus
berumur panjang, daripada sel-sel penguat transit yang
berumur pendek, dapat mengarah pada transformasi mereka,
menunjukkan bahwa sel-sel induk adalah sel-asal-asal pada
kanker usus [42]. Selain itu, kultur jangka panjang juga dapat
menginduksi sel batang mesenkim manusia dewasa transduksi
telomerase (hMSCs) untuk menjalani transformasi spontan, hanyamerekajaringan-spesifiksel-selstem tetapi juga sel induk
menunjukkan bahwa sel-sel ini juga merupakan asal dari CSCs jaringan lainnya. Misalnya, sel-sel sumsum yang diturunkan
[43, 44]. Menariknya, CSC berasal dari transformasi tidak tulang (BMDCs) mungkin menjadi penting
2 Stem Sel Internasional

dedifferentiation

Mutasi, onkogenik
aktivasi

Self-
pembaharuan
Self- penghambatan
pembaharuan

erentiati Apoptosis / sel


siklus penangkapan
DCSC
edifferentiati
edifferentiati
on
on
edifferentiati

on
Diferensiasi

Massal tumor

Gambar 1: Asal usul CSC dan terapi kombinasi penargetan CSC dan ablasi tumor curah. CSC mungkin berasal dari salah satu sel dengan mutasi / onkog
yang telah mengalami mutasi, atau daridistem.ffsel erentiatedatau sel-sel kanker yang diperoleh sifat batang-seperti oleh dediffdiferensiasi Dengan demikia
disarankan bahwa terapi kombinasional penargetan CSC dan curah ablasi tumor mungkin memilikieterapi yang lebih baikffEctsuntuk meningkatkan hasil kl
Sel induk
pembaharuan diri
CSC dari masing-masing jenis kanker tercantum pada Tabel 1.

3. Mekanisme yang olehnya CSC


Berkontribusi pada Perlawanan
terhadap Kemoterapi dan Kambuhnya
Kanker

Penelitian terbaru menunjukkan bahwa CSC diperkaya setelah


kemoterapi karena sedikit subpopulasi sel yang tersisa dalam
jaringan tumor, yang disebut CSCs, dapat bertahan dan
berkembang meskipun sebagian besar agen kemoterapi
membunuh sebagian besar tumor [12-14]. Misalnya,
preleukemic DNMT3Amut HSCS yang dapat melakukan
Sumberdari banyak jenis tumor, seperti kanker lambung, ekspansi klonal sebagai pertamalangkah dalam
tumor saraf, dan bahkan teratoma [45]. leukemogenesis dan meregenerasi seluruh hirarki
CSC juga telah dibuktikan yang akan dihasilkan hematopoietik ditemukan untuk bertahan hidup dan
olehdediffdiferensiasidari sel progenitor atau differentiated sel berkembang dalam remisi sumsum tulang setelah kemoterapi
yang telah diperoleh “stemness” sifat sebagai akibat dari [54]. Demikian pula, paparan dosis terapi temozolomide
akumulasi tambahan genetik atau epigenetik malities abnor- (TMZ), kemoterapi antiglioma yang paling umum digunakan,
[46]. Sebagai contoh, protein fusi BCR-ABL hadir dalam sel secara konsisten memperluas kumpulan sel induk glioma
induk hematopoietik - (HSC-) seperti sel CML tetapi nenek (GSC) dari waktu ke waktu dalam garis sel glioma yang
moyang granulosit-makrofag ditemukan menjadi kandidat berasal dari pasien dan yang telah terbentuk, yang telah
LSC stadium lanjut selama krisis ledakan di CML ledakan- terbukti merupakan hasil interkonversi fenotipik dan
krisis dengan mengaktifkan proses pembaharuan diri melalui fungsional antara differentiated sel tumor dan GSCs [55].
βjalur-catenin [47]. Selain itu, telah ditunjukkan bahwa Selain itu, antibodi VEGF yang dimanusiakan bevacizumab
pensinyalan Hh onkogenik dapat mempromosikan mengurangi pertumbuhan tumor glioblasoma multiforme
medulloblasoma baik dari nenek moyang sel granula atau sel (GBM) tetapi diikuti oleh kekambuhan tumor, mungkin karena
punca [48, 49]. Selain itu, kebanyakan differentiated sel-sel di penguncian autokrin yang sedang berlangsung melalui sumbu
SSP, termasuk parah differentiated neuron dan astrosit, dapat VEGF-VEGFR2-Neuropilin-1 (NRP1), yang terkait dengan
memperoleh defiperubahan genetik ned untuk dedif- ferentiate pengayaan bagian aktif VEGFR2 GSC dalam sel GBM
ke NSC atau negara progenitor dan akibatnya mendorong dan manusia [56]. The gefitinib tahan subline HCC827-GR-
memelihara glioma ganas [50]. tertinggi yang ditetapkan oleh metode-konsentrasi tinggi juga
Dari catatan, CSC memperoleh baik EMT dan batang fitur seperti sel tetapi tidak
dapatdiidentifikasiolehspesifikpenanda sel induk normal yang menunjukkan EGFR-mutan- spesifikfiproduksic proteinatau
biasa digunakan untuk mengisolasi CSC dari tumor padat dan peningkatan lebih lanjut dalam jumlah baik alel mutan atau
hematologi [51]. Beberapa penanda permukaan sel salinan EGFR [57]. Oleh karena itu, dengan memahami
telahdiverifikasiuntuk mengidentifikasi CSC- diperkaya mekanisme dan driver onkogenik dimana CSC melarikan diri
populasi, seperti CD133, CD24, CD44, EpCAM (molekul radio dan kemoterapi, kita dapat mengembangkan lebih
adhesi sel epitel), THY1, ABCB5 (ATP-mengikat kaset B5), effperawatan efektif yang bisa meningkatkan hasil klinis
dan CD200 [27, 32, 34, 52]. Selain itu, protein intraseluler pasien kanker. Mekanisme kontribusi CSC terhadap
tertentu juga telah digunakan sebagai penanda CSC, seperti kemoresisten termasuk EMT, MDR, dormansi, lingkungan
aldehyde dehydrogenase 1 (ALDH1) yang digunakan untuk tumor, dan sebagainya disebutkan di bawah ini secara
mengkarakterisasi CSCs dalam banyak jenis kanker seperti terperinci dan dirangkum dalam Gambar 2.
leukemia, payudara, usus besar, hati, paru-paru, pankreas, dan
sebagainya [ 12, 53]. Penggunaan penanda permukaan sel 3.1. Epithelial Mesenchymal Transition (EMT). Telah
sebagai CSC penanda kekuatan differ dari masing-masing jenis diindikasikan bahwa penanda transisi mesenchymal (EMT)
kanker tergantung pada karakteristik dan fenotipe mereka. epitel dan penanda sel punca secara bersamaan diekspresikan
Penanda permukaan yang sering digunakan untuk mengisolasi dalam sirkulasi sel tumor dari pasien dengan metastasis [58]
dan induksi EMT atau aktivasi faktor transkripsi EMT (TF) sel-sel yang telah mengalami EMT, dan EMT menginduksi
menganugerahkan fitur seperti batang pada sel kanker [ 59]. generasi angka yang relatif terbatas sel induk kanker dari lebih
Dalam sel-sel induk-seperti payudara manusia tertentu, normal differentiated sel neoplastik
dan neoplastik mengekspresikan penanda yang sama dengan
3 Stem Sel Internasional

Tabel 1: Kanker penanda sel induk pada manusia.

Jenis tumor Penanda sel batang kanker Referensi Kanker paru CD133 +, CD44+, ABCG2, ALDH, CD87+, SP, CD90+ [215-217] Kanker usus besar CD133+,
CD166+, EpCAM+, ALDH, ESA [218, 219] Kanker hati CD133+, CD44+, CD49f+, CD90+, ALDH, ABCG2, CD24+, ESA [51, 219] Kanker payudara CD13
EpCAM+, ALDH-1 [51, 218] Kanker lambung CD133+, CD44+, CD24+ [215, 220-222] Leukemia (AML) CD34+, CD38-, CD123+ [216, 218, 223] Kanker p
α2β1, ABCG2, ALDH [51, 215, 223] Kanker pankreas CD133 +, CD44+, CD24+, ABCG2, ALDH, EpCAM+, ESA [195, 215, 218] Melanoma ABCB5+, CD
kepala dan leher SSEA-1+, CD44+, CD133+ [224-226]
[60]. Sementara itu, ada hubungan antara aktivasi EMT dan resistensi obat [61]. Sebagai contoh,gefisel-sel kanker paru resisten yang d
memperoleh fenotipe EMT [62] melalui aktivasi pensinyalan Notch-1 [63]. Selain itu, potensi invasif yang ditingkatkan, tumorigenis
penanda EMT dapat digunakan untuk memprediksi resistensi cetuximab antibodi anti-EGFR dalam sel [64]. Secara paralel, dibanding
sel epitel, sel-sel mesenchymal telah meningkatkan ekspresi gen yang terlibat dalam metastasis dan invasi dan secarasignifikanlebi
penghambatan EGFR, termasuk erlotinib, gefitinib, dan cetuximab; setidaknya sebagian melalui overekspresi integrin-linked kinase
mesenkim [65]. Selain itu, mediator EMT S100A4 telah terbukti terlibat dalam mempertahankan sifat batang dan tumorigenicity CS
leher kanker [66]. Oleh karena itu, EMT menginduksi sel kanker untuk menunjukkan karakteristik seperti sel punca yang memprom
menyerang jaringan di sekitarnya dan menampilkan resistensi terapeutik [67]. Menariknya, ZEB1, regulator EMT, memainkan peran p
kunci sel induk kanker termasuk regulasi batang dan induksi chemoresistance melalui regulasi transkripsi O-6-Methylguanine DNA M
(MGMT) via miR-200c dan c-MYB pada glioma ganas [68]. Terlepas dari EMT, ekspresi tinggi penanda sel induk seperti Oct4, Nanog
dan Lgr5 telah dianggap untuk memberikan sistance chemore- serta[69-73].
3.2. Tingkat Tinggi Perlawanan Multidrug (MDR) atau DetoxifiProteinkasi. Sel-sel populasi samping (SP), yang menunjukkan fenotip
kanker, terdeteksi dalam
4 Stem Cells International
Resistant terhadapdipicu kerusakan DNA.
kematian sel yangMeningkatkan pemulungan ROS Mempromosikan kemampuan perbaikan DNA Mengaktifkan jalur pensinyalan anti-apoptosis
Gambar 2 : jalur sinyal Key danmodifikationdari CSC berkontribusi terhadap perlawanan terhadap kemoterapi. Dalam rangka untuk bertahan hidup
terapi, CSC menampilkan banyak tanggapan termasuk EMT, pembaruan diri, lingkungan tumor, ketenangan, epigenetikyang,modifikasi MDR
Mekanisme kontribusi CSC terhadap resistensi terhadap terapi dirangkum.

Lingkungan tumor (i)


Hipoksia (ii)
Autophagy (iii)
Kanker broblast (CAFs) yang berhubungan dengan kanker Peradangan Sel imun
Mekanisme epigenetik (i)
Modifikasi histone (ii) Metilasi DNA
Menjalani induksi EMT EMT atau aktivasi EMT-faktor transkripsi
(i)
(ii)
(i) (ii)
G1 / S
Kemampuan pembaruan diri
G0
(iv) (v)
G2 / M CSC Diam
Jalur sinyal
Wntpa
Jalur landak
CSC
Ekspresi yang lebih tinggi resistensi multidrug
(MDR) atau detoksifikasi protein Obat-transporter protein: ABCG1, ABCB1 Aldehid dehidrogenase (ALDH)
MRP
berbagai different tumor padat seperti retinoblastoma, roblastoma neutrofil, kanker pencernaan, kanker payudara, kanker paru-paru,
ekspresi tinggi protein pengangkut obat (termasuk MDR1, ABCG2, dan ABCB1) tidak hanya bertindak untuk mengecualikan pewar
juga mengeluarkan obat sitotoksik, yang mengarah pada resistensi tinggi terhadap agen kemoterapi dengan kelangsungan hidup sel ya
kekambuhan penyakit [74-76] ] Alisi et al. menunjukkan bahwa ekspresi berlebih dari protein ABC mungkin merupakan mekanisme p
paling penting untuk CSCs dalam menanggapi agen kemoterapi. Menariknya, telah menunjukkan bahwa PI3K / Aktsecarakhusus m
ABCG2 melalui lokalisasi untuk membran plasma, dan kehilangan PTEN pro motes fenotip SP kanker glioma batang-seperti sel ma
itu, aktivitas aldehida dehidrogenase (ALDH), enzim sitosolik yang bertanggung jawab untuk tion oxida- dari aldehida intraseluler
sel-sel darisecara potensial beracun effEctspeningkatan kadar spesies oksigen reaktif (ROS) [79] , adalah tinggi di kedua normal da
CD38- sel induk leukemia, dan dengan demikian memainkan peran penting dalam perlawanan terhadap kemoterapi [80]. Kegiata
penanda selektif esensial poten- untuk sel induk kanker di berbagaidiffjeniserent kanker, seperti kanker payudara [53], kanker kan
kepala dan leher skuamosa karsinoma sel [82], kanker paru-paru [83 ], dan rhabdomyosarcoma embrional [84]. Antar estingly, model
dan sampel klinis ies stud- menunjukkan bahwa sel-sel induk kanker ALDH1A1-positif mempromosikansignifikanketahanan untuk k
(gefitinib) dan obat antikanker kemoterapi lainnya (cisplatin, etoposid, dan fluorouracil ) dibandingkan sel-sel ALDH1A1-negatif pada
[85]. Selain itu, tingkat ekspresi ALDH1 yang tinggi
respon atau resistensi yang buruk terhadap terapi dengan meningkatkan pembersihan ROS, promosi kemampuan
kemoradioaktif pra operasi pada pasien kanker kerongkongan perbaikan DNA melalui ATM dan fosforilasi CHK1 / CHK2,
yang dapat direseksi [86]. atau aktivasi jalur pensinyalan anti-apoptosis, seperti PI3K /
Akt, WNT / b-catenin, dan Notch jalur pensinyalan [24].
Misalnya, CD44, molekul adhesi yang diekspresikan dalam
3.3. Dormansi CSC. Telah dibuktikan bahwa selain
CSC, berinteraksi dengan transporter glutamat-sistin dan
heterogenitas intratumoral yang diprakarsai oleh evolusi
mengendalikan level intraseluler dari glutathione tereduksi
subklon beragam secara genetik, ada juga klon yang berbeda
(GSH); karenanya, CSC yang mengekspresikan CD44 tingkat
secara fungsional, yang ditemukan dengan melacak keturunan
tinggi menunjukkan peningkatan kapasitas untuk sintesis GSH,
sel tunggal menggunakan lentivirus, dalam garis keturunan
menghasilkan pertahanan yang lebih kuat terhadap ROS [89].
genetik pada kanker kolonal [ 87]; dengan demikian, proses-
Menariknya, mirip dengan sel-sel induk jaringan normal,
proses yang menghasilkan keragaman dalam klon genetik ini
CSCs memiliki tingkat ROS yang lebih rendah, yang dikaitkan
mempromosikan sel-sel untuk potensi kelangsungan hidup
dengan peningkatan ekspresi sistem pembilasan radikal bebas,
yang lebih tinggi, terutama selama stres seperti kemoterapi.
yang mengarah ke pertahanan ROS yang lebih tinggi dan
Sebagai contoh, kemoterapi dapat menginduksi pertumbuhan + -/ rendah
tumor dari garis silsilah yang sebelumnya relatif tidak aktif resistensi radioterapi juga [90]. Selain itu,CD44 / CD24
atau berkembang biak yang masih mempertahankan potensi subset CSCpada kanker payudara resisten terhadap radiasi
penyebaran tumor yang kuat, yang mengarah pada melalui aktivasi pensinyalan ATM tetapi tidak tergantung pada
pertumbuhan kanker dan resistensi obat [87]. Demikian pula, perubahan aktivitas perbaikan DNA nonhomologous end
dalam glioblastoma multiforme, ada juga keberadaan subset bergabung (NHEJ) [91]. Demikian pula, CD133-
sel tumor endogen yang relatif diam dengan karakteristik yang mengekspresikan sel tumor yang diisolasi dari kedua
mirip dengan sel punca kanker yang bertanggung jawab untuk xenografts glioma manusia dan pri-mary spesimen pasien
mempertahankan pertumbuhan tumor jangka panjang dan oleh glioblastoma istimewa mengaktifkan pos pemeriksaan
karena itu menyebabkan kekambuhan melalui produksi kerusakan DNA dalam menanggapi radiasi dan perbaikan
populasi sementara yang sangat proliferatif. sel [17]. Secara radiasi kerusakan DNA lebih effectively dari sel-sel tumor
bersamaan, kerusakan yang diinduksi kemoterapi merekrut CD133-negatif [92] . Notch pathway juga mempromosikan
kelompok CSC kandung kemih yang bertahan lama selama radioresistensi sel punca glioma karena penghambatan Notch
periode celah antara siklus kemoterapi menjadi respons dengan gamma-secretase inhibitor (GSIs) menginduksi sel
pembelahan sel yang tak terduga untuk mengisi kembali tumor punca glioma agar lebih peka terhadap radiasi pada dosis yang
residual, mirip dengan mobilisasi perbaikan luka sel-sel batang relevan secara klinis karena pengurangan PI3K yang diinduksi
penduduk jaringan [88 ] oleh radiasi. / Aktivasi Akt dan peningkatan
level isoform apoptosis terpotong dari Mcl-1 (Mcl-1)
sementara tidak mengubah respon kerusakan DNA [93].
3.4. Ketahanan terhadap kematian sel yang disebabkan oleh
kerusakan DNA. CSC dapat resisten terhadap kematian sel
3.5. Lingkungan Tumor. Telah ditunjukkan bahwa lingkungan
yang disebabkan oleh kerusakan DNA melalui beberapa cara.
mikro yang berbeda dari berbagai komposisi seluler adalah
Ini termasuk perlindungan terhadap kerusakan DNA oksidatif
penting untuk melindungi dan mengatur sel-sel induk normal. tahan CSC paru-paru pada mutasi EGFR positif NSCLC oleh
Lingkungan mikro yang setara juga ditemukan dalam CSCs di upregulating ekspresi insulin-like growth factor 1 (IGF1)
mana CSCs didukung dengan baik dalam ceruk histologis, melaluiHIF1α dan mengaktivasi vating IGF1 reseptor (IGF1R)
yang disebut lingkungan mikro CSC [94-96], yang [118]. Menariknya, autophagy diregulasi dalam kanker
mengandung stroma konektif [97-101] dan jaringan pembuluh pankreas pada kondisi mental microenviron- oksigen rendah
darah [102-106]. Lingkungan ini mempercepat CSC dinamika dan kekurangan gizi, mirip dengan tumor hipoksia, dan
divisi, allow- ing mereka untukdiffsel-sel progenitor erentiate kemudian mempromosikan kelangsungan hidup clonogenic
anakserta memperbaharui diri dan mempertahankan CSC di dan migrasi dari pankreas CSCyang tinggi sel[119].
kondisi mental mengembangkan- primitif. Sel-sel dalam
lingkungan mikro CSC mampu menstimulasi jalur pensinyalan 3.5.2. Cancer-Associated Fibroblasts (CAFs). Sudah puncak-
[58], seperti Notch [102, 107, 108] dan Wnt [109-111] yang kasikan bahwa selain ketahanan otonom sel CSC, motherapy
dapat memfasilitasi CSC untuk bermetastasis, menghindari che- istimewa menargetkan non-CSC oleh stimulasi terkait
anoikis, dan mengubah dinamika divisi, mencapai repopulasi kanker fibroblasts (CAFS) yang menciptakan ceruk
dengan pembagian simetris [109, 112-114]. chemoresistant oleh peningkatan sekresi dengan spesialisasi
cific sitokin dan kemokin, termasuk interleukin-17A (IL-17A),
3.5.1. Hipoksia. Jalur pensinyalan Hypoxia dan HIF telah faktor pemeliharaan CSC dengan mempromosikan pembaruan
terbukti berkontribusi pada regulasi dan keberlanjutan CSC diri dan invasi [120]. Telah ditunjukkan sebelumnya bahwa
dan fenotip EMT seperti migrasi sel, invasi, dan angiogenesis CSC bisa differentiate ke dalam sel CAF-seperti (CAFLCs)
[115], melalui peningkatan ekspresi gen tanda tangan VEGF, dan karenanya mereka adalah salah satu sumber utama CAFS
IL-6, dan CSC seperti Nanog, Oct4, dan EZH2, pada kanker yang mendukung pemeliharaan tumor dan kelangsungan hidup
pankreas misalnya [116]. Oleh karena itu, jalur pensinyalan di relung kanker [121]. CAFS diketahui mengeluarkan
HIF juga dapat berperan dalam resistensi CSC terhadap terapi. banyakdifffaktor pertumbuhanerent, sitokin, dan kemokin,
Dalam lingkungan mikro hipoksia, faktor yang diinduksi termasuk faktor pertumbuhan tocyte hepa- (HGF), yang
hipoksia dan faktor hipoksia HIF1-α meningkatkan persistensi mengaktifkan tor MET recep- untuk melindungi CSC dari
sel CML terutama melalui upregulasi faktor yang diinduksi apoptosis dalam menanggapi monoterapi cetuximab
hipoksia 1α (HIF1-α), independen dari aktivitas kinase BCR- menargetkan EGFR di kanker kolorektal metastatik [122].
ABL1 [ 117]. Demikian pula, hipoksia meningkatkan gefitinib
5 Stem Cells International
3.5.3.Peradangan.Selain itu, pengobatan jangka panjang sel terhadap kemoterapi dengan mengganggu interaksi sel stroma /
kanker payudara dengan trastuzumabsecarakhusus diperkaya leukemia dalam lingkungan mikro sumsum tulang oleh Akt
CSC yang menunjukkan fenotipe EMT dengan tingkat yang phosphoryla- penghambatan tion dan induksi pembelahan
lebih tinggi dari sitokin yang disekresi IL-6 dibandingkan PARP karena bortomomib di hadapan sel stroma sumsum
dengan sel-sel tua; sebagai akibatnya, sel-sel ini tulang (BMSCs) dalam coculture [128]. Selain itu, CSC dari
mengembangkan resistensi trastuzumab dimediasi oleh tumor chemoresistant memiliki kemampuan unik untuk pro
aktivasi dari IL-6-dimediasi diflumpan balik inflamasi untuk Duce berbagai proinperadangan,sinyal seperti IFN peraturan
memperluas populasi CSC [123]. Demikian pula,TGF- faktor-5 (IRF5), yang bertindak sebagai faktor
autokrinβ pensinyalandan ekspresi IL-8 juga ditingkatkan transkripsispesifikuntuk tumor chemoresistant untuk
setelah pengobatan kemlitaxel obat kemoterapi pada kanker menginduksi Produksi M-CSF, untuk menghasilkan sel
payudara triple-negative, yang mengarah pada pengayaan myeloid imunoregulatori M2-like dari CD14+ monosit, dan
populasi CSC dan kekambuhan tumor [124]. Lebih lanjut, untuk mempromosikan aktivitas tumorigenik dan sel punca
kemokin stroma yang diturunkan faktor stroma yang yang dimediasi sel myeloid dari tumor massal [129].
diturunkan 1a (SDF-1a) dan reseptor serumpunnya CXCR4
memainkan peran penting dalam migrasi sel hematopoietik ke 3.5.4. Sel Kekebalan Tubuh. Telah diindikasikan sebelumnya
lingkungan mikro sumsum tulang [125, 126], jadi SDF-1A / bahwa makrofag terkait-tumor (TAMs) dapat mempromosikan
CXCR4 interaksi memediasi resistensi sel leukemia terhadap kemoresistensi pada kedua garis sel myeloma dan sel myeloma
apoptosis yang diinduksi kemoterapi [127], dan dengan primer dari apoptosis yang diinduksi oleh obat kemoterapi atau
demikian penghambatan CXCR4 dengan inhibitor seperti obat kemoterapi secara langsung dengan berinteraksi dengan
AMD3100 dapat meningkatkan sensitivitas sel leukemia sel-sel ganas dalam lingkungan mikro tumor dan melemahkan
aktivasi dan pembelahan dari pensinyalan apoptosis yang dalam berbagai jenis kanker, termasuk karsinoma nasofaring
tergantung caspase [130]. Selain itu, TAM juga secara [136], melanoma [137], adenokarsinoma pankreas [138],
langsung menginduksi sifat CSC sel tumor pankreas dengan kanker ovarium [139], dan karsinoma hepatoselular [140].
mengaktifkan transduser sinyal dan aktivator transkripsi 3 Lebih jauh lagi, anggota PcG lainnya EZH2, subunit katalitik
(STAT3) dan dengan demikian menghambatantitumor CD8 + dari kompleks penekan polycomb 2 (PRC2) yang memetilasi
respon limfosit TT dalam respon kemoterapi [131]. Selain itu, histon H3 pada lisin 27 (H3K27me) dan memunculkan
pada adenokarsinoma duktus pankreas, sel kanker mensekresi pembungkaman gen, juga berpartisipasi dalam kemoterapi
faktor penstimulasi koloni 1 (CSF1) untuk menarik dan kanker kanker pankreas dengan membungkam gen penekan
menstimulasi reseptor CSF- (CSF1R-) yang mengekspresikan tumor p27 melalui metilasi histone H3 -lysine 27 (H3K27)
TAM untuk mengekspresikan tingkat tinggi sitidin deaminase [141]. Selain itu, EZH2 melindungi GSC dari kematian sel
(CDA), suatu enzim intraseluler yang mengkatalisasi enzim yang diinduksi radiasi dan akibatnya mempromosikan
bentuk bioaktif dari tabibin dan oleh karena itu melindungi sel-kelangsungan hidup GSC dan radioresisten melalui regulator
sel kanker dari kemoterapi [132]. hulu mitosis kinase embrionik leucine-ritsleting kinase ibu
(MELK) [142]. Selain itu, penghambatan EZH2 peka BRG1
3.6. Epigenetik. Selain itu, resistensi obat CSC-dimediasi dan EGFR hilangnya fungsi tumor mutan untuk topoisomerase
diatur oleh mekanisme epigenetik juga, termasuk histone II (TopoII) inhibitor etoposide dengan peningkatan fase S,
modifikasi dan metilasi DNA. Pertama, DNA metilasi tidak bridging anafase, dan apoptosis [143]. Menariknya, EZH2 dan
berubah selama TGF-ßEMT -dimediasi tetapi perubahan BMI1 diindikasikan berkorelasi terbalik dengan tanda tangan
epigenetik lain seperti lysine-spesifikdeaminase-1- prognosis dan mutasi TP53 pada kanker payudara [144].
pengurangan global (Lsd1-) tergantung Kedua, asetilasi histone terlibat dalam regulasi aktivasi
dari tanda heterochromatin H3-lys9 dimethylation transkripsi dan chemoresistance dari CSC juga. Pengobatan
(H3K9Me2), meningkat dari tanda euchromatin H3-lys4 dengan HDAC inhibitor (HDACi) effectively menargetkan
trimethylation (H3K4Me3) dan tanda transkripsi H3-lys36 diam kronis myelogenous Kemia leu- (CML) stem sel yang
trimethylation (H3K36Me3) ditemukan; terutama, H3K4Me3 tahan terhadap tyrosine kinase inhibitor imatinib mesylate
mungkin berkontribusi terhadap chemoresistance yang diatur (IM) [145]. Demikian pula, pretreatment dengan inhibitor
Lsd1 [133]. Selain itu, KDM1A, sebuah flavin yang me- HDAC dapat membuat peka sel-sel seperti batang prostat
sembilan dinucleotide- (FAD-) tergantunglysine- untuk pengobatan radiasi melalui peningkatan kerusakan DNA
spesifikdemethylasesecarakhusus dengan monomethyl- dan dan mengurangi kelangsungan hidup klonogenik [146].
dimethyl- histon H3 lisin-4 (H3K4) dan lisin-9 (H3K9) sub Vorinostat, inhibitor HDAC melalui merangsang tination
Strate, merupakan regulator penting dari MLL-Af9 sel ubiqui- dan degradasi lisosom, downregulates ekspresi dan
leukemia induk (LSC) potensi onkogenik dengan menghalangi signaling dari ketiga reseptor EGFR, ErbB2, dan ErbB3
differenti- asi [134]. Selain itu, B-cell-spesifikfiintegrasic bersama-sama dengan pengembalian EMT di EGFR TKI
Moloney murine leuke- mia virus situs 1 (BMI1), salah satu gefisel tinib-tahan dan karena itu meningkatkan antitumor
dari beberapa protein peredam epigenetik milik kelompok effect gefitinib dalam karsinoma sel skuamosa kepala dan leher
Polycomb (PCG), diperlukan untuk pembaruan diri dari kedua [147]. Menariknya, NANOG meningkatkan pengaturan
sel induk dewasa dan banyak CSC melalui berbagai jalur histone deacetylases 1 (HDAC1) melalui pengikatan ke daerah
utama, seperti pertumbuhan independen-pelabuhan, jalur Wnt promotor dan mengurangi asetilasi H3 nada K14 dan K27;
dan Notch [135]. BMI-1 telah diindikasikan terlibat dalam sebagai hasilnya, ia menginduksi tidak hanya fitur seperti
perlindungan sel kanker dari apoptosis atau resistensi obat batang melalui represi epigenetik sel. Sel
6 Stem SelInternasional
Stem Sel Internasional 7
Aktivasi penghambat siklus CDKN2D dan CDKN1B tetapi juga dari Wnt /β-catenin dan ATP-mengikat k
imun dan chemoresistance me
(ABCG2) pada kanker ovarium [164]. upregulation by epigenetic silencing of E3 ubiquitin-ligase
Secondly, Hh pathway could regulate autophagy in CML TRIM17 and NOXA [148].
cells and then inhibition of the Hh pathway and autophagy Third, many tumor suppressor genes have been shown to
simultaneously could sharply reduce cell viability and signif- be epigenetically silenced in chemoresistant cancers by DNA
icantly induce apoptosis of imatinib-sensitive or -resistant methylation on CpG promoter regions. For instance, tumor
BCR-ABL+ cells via downregulating the kinase activity of suppressor insulin-like growth factor binding protein-3
the BCR-ABL oncoprotein [165]. Concomitantly, the expres- (IGFBP-3), which is involved in controlling cell growth,
sion of sonic hedgehog (SHH) and glioma-associated onco- transformation, and survival, is specifically downregulated
gene homolog 1 (GLI1), the well-known signaling pathway through promoter-hypermethylation and results in acquired
molecules involved in the drug resistance, is higher in resistance to chemotherapy in many different types of cancer
enriched CD44+/Musashi-1+ gastric cancer stem cells and [149]. In addition, loss of DNA mismatch repair (MMR)
consequently enhances the drug resistance via high drug gene hMLH1 via full hypermethylation of the hMLH1 pro-
efflux pump activity [166]. In glioma, CD133 + CSC popula- moter [150] is highly correlated with the ability of arresting
tion, which contributes to the chemoresistance of therapy cell death and cell cycle after DNA damage induced by che-
such as temozolomide (TMZ) treatment, overexpresses genes motherapy and poor survival prediction for cancer patients
involved in Notch and SHH pathways and activates these [151], hence plays a role in drug resistance in ovarian [152]
pathways [167]. and breast cancers [153].
Last but not least, chemotherapy such as oxaliplatin induces Notch-1 receptor and its downstream target Hes-1 3.7. Signaling Pathway
Che
activity by increasing gamma-secretase activity in colon can- mentioned, normal stem cells and CSCs have similar charac-
cer cells; hence, inhibition of Notch-1 signaling by gamma- teristics such as self-renewal and differentiation. They also
secretase inhibitors (GSIs) sensitizes colon cancer cells to share numbers of key signaling pathway to maintain its exis-
chemotherapy [168]. Moreover, Notch signaling pathway tence. For example, Notch signaling was highly expressed in
and Notch3 in particular play an essential role in the regula- the hematopoietic tumors such as T-ALL and solid tumors
tion of CSC maintenance and chemoresistance to platinum such as non-small-cell lung carcinoma (NSCLC), breast can-
in ovarian cancer therapy [169]. Similarly, the enrichment cer, and glioblastoma [154–156]. Activation of Hedgehog sig-
of CD133+ cells in lung adenocarcinoma after cisplatin naling which in normal condition plays important roles in
induction leads to multidrug resistance through activation embryonic development and tissue regeneration also has
of Notch signaling as higher levels of cleaved Notch1 been found to be involved in the regulation of various cancer
(NICD1) are detected [170]. Furthermore, it has been shown stem cells, such as pancreatic cancer, leukemias, and basal
that gefitinib-acquired resistant lung adenocarcinoma cells cell carcinoma (BCC) [157]. Another signaling pathway such
undergo EMT by activation of Notch-1 signaling via as WNT, TGFβ, PI3K/Akt, EGFR, and JAK/STAT, as well as
Notch-1 receptor intracellular domain (N1IC), the activated transcriptional regulators including OCT4, Nanog, YAP/
form of the Notch-1 receptor [63]. TAZ, and Myc are also commonly activated in various cancer
Besides, there are also some molecules which act as the stem cells to regulate their self-renewal and differentiation
integration of various pathways involved in the control of state [21, 158]. CSCs have been indicated to display many
stem cell fate across tissues; for example, CYP26, a primary characteristics of embryonic or tissue stem cells and develop-
retinoid-inactivating enzyme through retinoid and Hedge- mental signaling pathways such as Wnt, HH, and Notch that
hog pathways, limits the retinoic acid concentration, there- are highly conserved embryonically and control self-renewal
fore leading to drug resistance in the stem cell niche [171]. of stem cells [159]. Therefore, activation of these pathways may play an imp
expansion of CSCs and hence the resistance to therapy [160]. Here, several represen-
4. CSC-Based Therapy
tatives are explained.
Owing to the ability of CSCs to develop chemo- and radiore- First, it has been indicated that activation of Wnt/β-
sistance which play key roles in the malignant progression, catenin signaling enhances the chemoresistance to IFN-α/5-
metastasis, and cancer recurrence, it is suggested that target- FU combination therapy [161]. OV6+ HCC cells, a subpopu-
ing cancer stem cells offers an ultimate goal to overcome a lation of less differentiated progenitor-like cells in HCC cell
poor prognosis, leading to a better patient survival [15, 22]. lines and primary HCC tissues, have been shown to be
Selective targeting of CSC signaling networks that are essen- endogenously active Wnt/β-catenin signaling and resistant
tial for self-renewal, proliferation, and differentiation to to standard chemotherapy [162]. In addition, in neuroblas-
maintain their stem cell properties provides a new challenge toma, amplification and upregulation of frizzled-1 Wnt
in the development of cancer treatments [19, 172]. Over the receptor (FZD1) activate the Wnt/β-catenin pathway in che-
last decades, it was suggested that the combination of con- moresistant cancer cells by nuclear β-catenin translocation
ventional therapy and targeted therapy against CSC-specific and transactivation of Wnt target genes such as multidrug
pathways gives rise a better consequence compared to mono- resistance gene (MDR1), which is known to mediate the
therapy in removal of both bulk tumor and CSC population resistance to chemotherapy [163]. Furthermore, c-Kit, a stem
(Figure 1) [19]. Thus, targeting essential pathways in the cell factor (SCF) receptor, mediates chemoresistance through
CSCs such as Notch, Wnt, and Hedgehog (HH) is being
developed to block the self-renewal of CSCs [21]. Lately, using microRNA [184–190].
some classes of Notch pathway inhibitors have been reported Accumulating evidence suggests that miRNA and
to enter a clinical trial, accompanied by a substantial variety of other groups of long noncoding RNA (lncRNA) play important
targets, mechanism of action, and drug classes [19, 21]. The roles in the regulation of CSCs properties such as self-renewal,
major class of Notch inhibitor is the γ-secretase inhibi- tors asymmetric cell division, tumor initiation, drug resistance, and
(GSIs). GSI works by inhibiting the final proteolytic cleavage disease recurrence [186, 187, 189, 191–193].The usage of
of Notch receptors, which results in the release of the active miRNA as CSC-based therapeutic agents is reported; for
intracellular fragment. It was the first class of Notch pathway example, mir-22 that targets TET2 in leukemia (AML and
inhibitor that enters a clinical trial in the can- cer field [159, MDS) and breast cancer [194], Let-7 to target RAS and
173, 174]. HMGA2 in breast cancer [195], mir-128 to target BMI-1 in
HH pathway is shown to be involved in several brain cancer [191], mir-200 to target ZEB1/ZEB2, BMI-1, and
essential developmental pathways such as tissue patterning SUZ12 in breast cancer [189, 196, 197], and some other
during embryonic development and the repair of normal miRNA in the colon cancer and prostate cancer have been
tissues and epithelial-to-mesenchymal transition [175]. reported to reduce cancer malignancy [198–202].
Vismode- gib, a drug targeting HH pathway, was approved by Finally, cancer immunotherapy may be a breakthrough
the Euro- pean Medicines Agency (EMA) in 2013 and the US for targeting specifically CSCs in cancer patients. For cancer
FDA in 2012 for the therapy of metastatic BCC patients or immunotherapy, several effectors, including natural killer (NK)
locally advanced BCC patients that are not candidates for cells and γδT cells in innate immunity, antibodies in acquired
surgery or radiotherapy [176, 177]. humoral immunity, CSC-based dendritic cells, and CSC-
Targeting Wnt signaling has also shown promising primed cytotoxic T lymphocytes (CTLs) in acquired cellular
results related to carcinogenesis, tumor invasiveness, and immunity, which are able to recognize and kill CSCs may be
metastasis [159]. Wnt3A-neutralizing mAb was shown to have suitable candidates to improve the efficacy of cancer treatment.
antiproliferation and proapoptotic effects in prostate cancer A variety of immunotherapeutic strategies that specifically
mouse model [178]. And anti-Fz10 radio-labeled mAb is being target CSCs using these effector cells have been reported. In
evaluated in a phase I trial for the synovial sarcoma therapy. addition, identification of specific antigens or genetic
Vantictumab (OMP-18R5, a mAb that blocks five Fz receptors alterations in CSCs plays an important role in finding targets
such as Fz1, Fz2, Fz5, Fz7, and Fz8) [179–181] and OMP- for immunotherapy. These include CSC markers (ALDH
54F28 [181] (a mAb that blocks fusion protein decoy receptor [203], CD44 [204, 205], CD133 [206], EpCAM [207], and
such as truncated Fz8) are under investigation in phase I HER2 [208]), CSC niche interaction (TAM [209]), tumor
studies in advanced-stage solid tumors [182]. microenvironment (immune cells/myeloid- derived suppressor
Targeting CSCs through the EMT pathways also cells), cytokines (IL1 [210], IL6 [211], and IL8 [212]), and
provides a new challenge in the cancer therapy study. This immune checkpoint (CTLA-4 [213] or PD1/PDL1 [214]).
therapy is developed in order to prevent cancer aggressiveness
and acquired drug resistance of cancer stem cells [183, 184]. 5. Conclusion
Lately, the finding of therapeutic agents to EMT-based CSC
therapy indicated three general target groups [184, 185]. These CSCs possess stem cell-like features found in cancer and have
include a group involved in the regulation of EMT important implications for the chemoresistance and cancer
extracellular inducer such as TGF-β, EGF, Axl-Gas6 path- relapse, a notion that remains somewhat controversial. With a
ways, hypoxia, and extracellular matrix components. Another small subpopulation in the malignant cell pool, the contri-
group is the transcription factors (TFs) that pro- mote EMT bution of CSCs is remarkable in cancer therapy, as shown by
transcriptome including Twist1, Snail1, Zeb1/2, T-box TF intensive studies in recent decades. These cells can be identi-
Brachyury as well as its downstream effectors of EMT, such as fied based on the presence of surface biomarkers, enhanced
E-Cadherin, N-Cadherin, vimentin, and HoxA9. The last one is spheroid or colony formation in vitro and augmented tumor-
targeting regulators of EMT-TFs and epigenetic regulator initiating potential as well as tumorigenic ability in vivo. They
are resistance to chemotherapy and radiation therapy compared
to bulk tumor cells and hence play a cru- cial role in tumor promising strategies for suppressing tumor relapse and
recurrence after anticancer therapy. To sur- vive following metastasis, recent technological advances made it easier than
cancer treatment, CSCs seem to be able to manifest several before to find mechanisms contributing to drug resistance.
responses such as EMT, induction of signal- ing pathways that Also, the recent therapeutic strategy of combining molecules
regulate self-renewal or influence tumor environments, specifically targeting CSCs with conventional che-
expression of drug transporters or detoxifica- tion proteins, and motherapeutic drugs could possibly be a better direction for
so forth to protect them from devastating effects caused by anticancer therapy and may therefore achieve better survival
therapeutic agents. Thus, the development of anticancer rates of cancer patients (Figure 1) [19]. Besides, as some cell
therapeutics that target CSCs is not only limited to the finding surface biomarkers and signaling pathways are similar
of inhibitor of CSC pathways and cell surface markers but also between CSCs and normal stem cells, it is also essentially
to the development of EMT and CSCs microenvironment- required to develop novel therapeutic agents targeting only
related inhibitors. Though the molecular mechanisms CSCs to avoid off-target effects on noncancerous cells or
underlying the resistance of CSCs to chemo- therapy and normal stem cells.
radiation still require further studies in order to develop
8 Stem Cells International
Conflicts of Interest no. 19, pp. 1441–1454, 2007. [8] DA Senthebane, A. Rowe, NE
Thomford et al., “The role of tumor microenvironment in
The authors declare no conflict of interest. chemoresistance: to survive, keep your enemies closer,”
International Journal of Molecular Sciences, vol. 18, tidak. 7, 2017.
[9] A. Fesler, S. Guo, H. Liu, N. Wu, and J. Ju, “Overcoming
Authors' Contributions chemoresistance in cancer stem cells with the help of microRNAs in
colorectal cancer,” Epigenomics, vol. 9, tidak. 6, pp. 793–796, 2017.
Lan Thi Hanh Phi and Ita Novita Sari contributed equally to [10] R. Di Fiore, R. Drago-Ferrante, F. Pentimalli et al., “Micro-
the work. RNA-29b-1 impairs in vitro cell proliferation, self‐ renewal and
chemoresistance of human osteosarcoma 3AB-OS cancer stem
cells,” International Journal of Oncology, vol. 45, no. 5, pp. 2013–
Acknowledgments 2023, 2014. [11] D. Chen, M. Wu, Y. Li et al., “Targeting BMI1+
cancer stem cells overcomes chemoresistance and inhibits
This work was supported by the Soonchunhyang University metastases in squamous cell carcinoma,” Cell Stem Cell, vol. 20,
Research Fund and Global Research Development Center tidak. 5, pp. 621–634.e6, 2017. [12] JE Visvader and GJ Lindeman,
“Cancer stem cells in solid tumours: accumulating evidence and
(NRF-2016K1A4A3914725).
unresolved questions,” Nature Reviews Cancer, vol. 8, tidak. 10, pp.
755–768, 2008.
[13] MR Alison, SM Lim, and LJ Nicholson, “Cancer stem cells:
References
problems for therapy?,” The Journal of Pathology, vol. 223, no. 2,
pp. 147–161, 2011. [14] M. Dean, T. Fojo, and S. Bates, “Tumour
[1] R. Siegel, D. Naishadham, and A. Jemal, “Cancer statistics,
stem cells and drug resistance,” Nature Reviews Cancer, vol. 5,
2013,” CA: a Cancer Journal for Clinicians, vol. 63, tidak. 1, pp. 11–
tidak. 4, pp. 275–284, 2005. [15] A. Singh and J. Settleman, “EMT,
30, 2013. [2] D. Hanahan and RA Weinberg, “Hallmarks of cancer:
cancer stem cells and drug resistance: an emerging axis of evil in the
the next generation,” Cell, vol. 144, no. 5, pp. 646–674, 2011. [3]
war on cancer,” Oncogene, vol. 29, tidak. 34, pp. 4741–4751, 2010.
YN You, VT Lakhani, and SA Wells Jr., “The role of pro- phylactic
[16] MH Kiyohara, C. Dillard, J. Tsui et al., “EMP2 is a novel
surgery in cancer prevention,” World Journal of Surgery, vol. 31,
therapeutic target for endometrial cancer stem cells,” Onco- gene,
tidak. 3, pp. 450–464, 2007. [4] BM Putzer, M. Solanki, and O.
vol. 36, tidak. 42, pp. 5793–5807, 2017. [17] J. Chen, Y. Li, TS Yu
Herchenroder, “Advances in cancer stem cell targeting: how to strike
et al., “A restricted cell population propagates glioblastoma growth
the evil at its root,” Advanced Drug Delivery Reviews, vol. 120, pp.
after chemotherapy,” Nature, vol. 488, no. 7412, pp. 522–526, 2012.
89–107, 2017. [5] YA Luqmani, “Mechanisms of drug resistance in
[18] D. Fang and H. Kitamura, “Cancer stem cells and epithe- lial–
cancer chemotherapy,” Medical Principles and Practice, vol. 14,
mesenchymal transition in urothelial carcinoma: possible pathways
tidak. 1, pp. 35–48, 2005. [6] PA Reid, P. Wilson, Y. Li, LG Marcu,
and potential therapeutic approaches,” International Journal of
and E. Bezak, “Current understanding of cancer stem cells: review of
Urology, vol. 25, tidak. 1, pp. 7– 17, 2018. [19] M. Cojoc, K.
their radiobiology and role in head and neck cancers,” Head & Neck,
Mäbert, MH Muders, and A. Dubrovska, “A role for cancer stem
vol. 39, no. 9, pp. 1920–1932, 2017. [7] O. Tredan, CM Galmarini,
cells in therapy resistance: cellular and molecular mechanisms,”
K. Patel, and IF Tannock, “Drug resistance and the solid tumor
Seminars in Cancer Biology, vol. 31, pp. 16–27, 2015. [20] LI
microenvironment,” Journal of the National Cancer Institute, vol. 99,
Shlush, A. Mitchell, L. Heisler et al., “Tracing the origins of relapse primitive hematopoietic cell,” Nature Medicine, vol. 3, tidak. 7, pp.
in acute myeloid leukaemia to stem cells,” Nature, vol. 547, no. 730– 7, 1997. [26] SA Joosse and K. Pantel, “Biologic challenges in
7661, pp. 104–108, 2017. [21] DR Pattabiraman and RA Weinberg, the detec- tion of circulating tumor cells,” Cancer Research, vol. 73,
“Tackling the cancer stem cells — what challenges do they pose?,” tidak. 1, pp. 8–11, 2013. [27] M. Al-Hajj, MS Wicha, A. Benito-
Nature Reviews Drug Discovery, vol. 13, no. 7, pp. 497–512, 2014. Hernandez, SJ Morrison, and MF Clarke, “Prospective identification
[22] ZJ Yang and RJ Wechsler-Reya, “Hit 'em where they live: of tumorigenic breast cancer cells,” Proceedings of the National
targeting the cancer stem cell niche,” Cancer Cell, vol. 11, no. 1, pp. Academy of Sciences of the United States of Amer- ica, vol. 100,
3–5, 2007. [23] P. Valent, D. Bonnet, R. de Maria et al., “Cancer no. 7, pp. 3983–3988, 2003. [28] TN Ignatova, VG Kukekov, ED
stem cell definitions and terminology: the devil is in the details,” Laywell, ON Suslov, FD Vrionis, and DA Steindler, “Human
Nature Reviews Cancer, vol. 12, tidak. 11, pp. 767–775, 2012. [24] cortical glial tumors contain neural stem-like cells expressing
C. Peitzsch, I. Kurth, L. Kunz-Schughart, M. Baumann, and A. astroglial and neuronal markers in vitro,” Glia, vol. 39, no. 3, pp.
Dubrovska, “Discovery of the cancer stem cell related determinants 193–206, 2002. [29] T. Schatton, GF Murphy, NY Frank et al.,
of radioresistance,” Radiotherapy and Oncol- ogy, vol. 108, no. 3, “Identification of cells initiating human melanomas,” Nature, vol.
pp. 378–387, 2013. [25] D. Bonnet and JE Dick, “Human acute 451, no. 7176, pp. 345–349, 2008.
myeloid leukemia is organized as a hierarchy that originates from a
9 Stem Cells International
10 Stem Cells International
intestinal cancer,” Nature, vol. 457, no. 7229, pp. 608–611, 2009.
[43] D. Rubio, J. Garcia-Castro, MC Martín et al., “Spontaneous
[30] SK Singh, ID Clarke, M. Terasaki et al., “Identification of a human adult stem cell transformation,” Cancer Research, vol. 65, no.
cancer stem cell in human brain tumors,” Cancer Research, vol. 63, 8, pp. 3035–3039, 2005. [44] JS Burns, BM Abdallah, P. Guldberg,
tidak. 18, pp. 5821–5828, 2003. [31] A. Eramo, F. Lotti, G. Sette et J. Rygaard, HD Schrøder, and M. Kassem, “Tumorigenic
al., “Identification and expan- sion of the tumorigenic lung cancer heterogeneity in cancer stem cells evolved from long-term cultures
stem cell population,” Cell Death & Differentiation, vol. 15, tidak. 3, of telomerase-immortalized human mesenchymal stem cells,”
pp. 504–514, 2008. [32] ZF Yang, DW Ho, MN Ng et al., Cancer Research, vol. 65, no. 8, pp. 3126–3135, 2005. [45] C. Liu,
“Significance of CD90+ cancer stem cells in human liver cancer,” Z. Chen, Z. Chen, T. Zhang, and Y. Lu, “Multiple tumor types may
Cancer Cell, vol. 13, no. 2, pp. 153–166, 2008. [33] C. Li, DG originate from bone marrow–derived cells,” Neoplasia, vol. 8, tidak.
Heidt, P. Dalerba et al., “Identification of pancre- atic cancer stem 9, pp. 716–724, 2006. [46] I. Baccelli and A. Trumpp, “The evolving
cells,” Cancer Research, vol. 67, no. 3, pp. 1030–1037, 2007. [34] concept of cancer and metastasis stem cells,” Journal of Cell
CA O'Brien, A. Pollett, S. Gallinger, and JE Dick, “A human colon Biology, vol. 198, no. 3, pp. 281–293, 2012. [47] CHM Jamieson,
cancer cell capable of initiating tumour growth in immunodeficient LE Ailles, SJ Dylla et al., “Granulocyte– macrophage progenitors as
mice,” Nature, vol. 445, no. 7123, pp. 106–110, 2007. [35] S. Zhang, candidate leukemic stem cells in
C. Balch, MW Chan et al., “Identification and characterization of blast-crisis CML,” The New England Journal of Medicine, vol. 351,
ovarian cancer-initiating cells from pri- mary human tumors,” no. 7, pp. 657–667, 2004. [48] U. Schüller, VM Heine, J. Mao et al.,
Cancer Research, vol. 68, tidak. 11, pp. 4311–4320, 2008. [36] TB “Acquisition of granule neuron precursor identity is a critical
Brunner, LA Kunz-Schughart, P. Grosse-Gehling, and M. Baumann, determinant of progen- itor cell competence to form Shh-induced
“Cancer stem cells as a predictive factor in radiotherapy,” Seminars medulloblastoma,” Cancer Cell, vol. 14, tidak. 2, pp. 123–134, 2008.
in Radiation Oncology, vol. 22, tidak. 2, pp. 151–174, 2012. [37] P. [49] ZJ Yang, T. Ellis, SL Markant et al., “Medulloblastoma can be
Dalerba, RW Cho, and MF Clarke, “Cancer stem cells: models and initiated by deletion of Patched in lineage-restricted progenitors or
concepts,” Annual Review of Medicine, vol. 58, no. 1, pp. 267–284, stem cells,” Cancer Cell, vol. 14, tidak. 2, pp. 135–145, 2008. [50]
2007. [38] M. López-Lázaro, “The migration ability of stem cells D. Friedmann-Morvinski, EA Bushong, E. Ke et al., “Dedif-
can explain the existence of cancer of unknown primary site. ferentiation of neurons and astrocytes by oncogenes can induce
Rethinking metastasis,” Oncoscience, vol. 2, tidak. 5, pp. 467– 475, gliomas in mice,” Science, vol. 338, no. 6110, pp. 1080–1084, 2012.
2015. [39] M. Lopez-Lazaro, “Stem cell division theory of cancer,” [51] K. Chen, Y.-h. Huang, and J.-l. Chen, “Understanding and
Cell targeting cancer stem cells: therapeutic implications and challenges,”
Cycle, vol. 14, tidak. 16, pp. 2547-2548, 2015. [40] Y. Acta Pharmacologica Sinica, vol. 34, tidak. 6, pp. 732–740, 2013.
Wang, J. Yang, H. Zheng et al., “Expression of mutant p53 proteins [52] P. Dalerba, SJ Dylla, IK Park et al., “Phenotypic character-
implicates a lineage relationship between neural stem cells and ization of human colorectal cancer stem cells,” Proceedings of the
malignant astrocytic glioma in a murine model,” Cancer Cell, vol. National Academy of Sciences of the United States of America, vol.
15, tidak. 6, pp. 514–526, 2009. [41] M. Nouri, J. Caradec, AA 104, tidak. 24, pp. 10158–10163, 2007. [53] C. Ginestier, MH Hur,
Lubik et al., “Therapy-induced developmental reprogramming of E. Charafe-Jauffret et al., “ALDH1 is a marker of normal and
prostate cancer cells and acquired therapy resistance,” Oncotarget, malignant human mammary stem cells and a predictor of poor
vol. 8, tidak. 12, pp. 18949–18967, 2017. [42] N. Barker, RA clinical outcome,” Cell Stem Cell, vol. 1, tidak. 5, pp. 555–567,
Ridgway, JH van Es et al., “Crypt stem cells as the cells-of-origin of 2007. [54] LI Shlush, S. Zandi, A. Mitchell et al., “Identification of
pre- leukaemic haematopoietic stem cells in acute leukaemia,” Brabletz, and J. Caramel, “Oncogenic roles of EMT-inducing
Nature, vol. 506, no. 7488, pp. 328–333, 2014. [55] B. Auffinger, transcription factors,” Nature Cell Biology, vol. 16, tidak. 6, pp.
AL Tobias, Y. Han et al., “Conversion of differ- entiated cancer cells 488–494, 2014. [60] SA Mani, W. Guo, MJ Liao et al., “The
into cancer stem-like cells in a glioblas- toma model after primary epithelial- mesenchymal transition generates cells with properties of
chemotherapy,” Cell Death & Differentiation, vol. 21, tidak. 7, pp. stem cells,” Cell, vol. 133, no. 4, pp. 704–715, 2008. [61] S.
1119–1131, 2014. [56] P. Hamerlik, JD Lathia, R. Rasmussen et al., Meidhof, S. Brabletz, W. Lehmann et al., “ZEB1‐ associated drug
“Autocrine VEGF–VEGFR2–Neuropilin-1 signaling promotes resistance in cancer cells is reversed by the class I HDAC inhibitor
glioma stem-like cell viability and tumor growth,” Journal of Experi- mocetinostat,” EMBO Molecular Medicine, vol. 7, tidak. 6, pp. 831–
mental Medicine, vol. 209, no. 3, pp. 507–520, 2012. [57] K. Shien, 847, 2015. [62] H. Uramoto, T. Iwata, T. Onitsuka, H. Shimokawa,
S. Toyooka, H. Yamamoto et al., “Acquired resis- tance to EGFR T. Hanagiri, and T. Oyama, “Epithelial−mesenchymal tran- sition in
inhibitors is associated with a manifestation of stem cell–like EGFR-TKI acquired resistant lung adenocarci- noma,” Anticancer
properties in cancer cells,” Cancer Research, vol. 73, tidak. 10, pp. Research, vol. 30, no. 7, pp. 2513–7, 2010. [63] M. Xie, L. Zhang,
3051–3061, 2013. [58] T. Oskarsson, E. Batlle, and J. Massague, CS He et al., “Activation of Notch-1 enhances epithelial–
“Metastatic stem cells: sources, niches, and vital pathways,” Cell mesenchymal transition in gefitinib-
Stem Cell, vol. 14, tidak. 3, pp. 306–321, 2014. [59] A. Puisieux, T.
11 Stem Cells International
characteristics in retinoblas- toma,” Molecular Vision, vol. 11, pp.
729–737, 2005. [75] C. Hirschmann-Jax, AE Foster, GG Wulf et al.,
acquired resistant lung cancer cells,” Journal of Cellular Bio- “A distinct “side population” of cells with high drug efflux capacity
chemistry, vol. 113, no. 5, pp. 1501–1513, 2012. [64] PC Black, GA in human tumor cells,” Proceedings of the National Academy of
Brown, T. Inamoto et al., “Sensitivity to epi- dermal growth factor Sciences of the United States of America, vol. 101, no. 39, pp.
receptor inhibitor requires E-cadherin expression in urothelial 14228–14233, 2004. [76] N. Haraguchi, T. Utsunomiya, H. Inoue et
carcinoma cells,” Clinical Cancer Research, vol. 14, tidak. 5, pp. al., “Characteriza- tion of a side population of cancer cells from
1478–1486, 2008. [65] BC Fuchs, T. Fujii, JD Dorfman et al., human gastro- intestinal system,” Stem Cells, vol. 24, tidak. 3, pp.
“Epithelial-to- mesenchymal transition and integrin-linked kinase 506–513, 2006. [77] A. Alisi, W. Cho, F. Locatelli, and D. Fruci,
mediate sensitivity to epidermal growth factor receptor inhibition in “Multidrug resistance and cancer stem cells in neuroblastoma and
human hepatoma cells,” Cancer Research, vol. 68, tidak. 7, pp. hepatoblastoma,” International Journal of Molecular Sci- ences, vol.
2391–2399, 2008. [66] JF Lo, CC Yu, SH Chiou et al., “The 14, tidak. 12, pp. 24706–24725, 2013. [78] AM Bleau, D.
epithelial- mesenchymal transition mediator S100A4 maintains Hambardzumyan, T. Ozawa et al., “PTEN/ PI3K/Akt pathway
cancer-initiating cells in head and neck cancers,” Cancer Research, regulates the side population phenotype
vol. 71, no. 5, pp. 1912–1923, 2011. [67] K. Polyak and RA and ABCG2 activity in glioma tumor stem-like cells,” Cell Stem
Weinberg, “Transitions between epithe- lial and mesenchymal states: Cell, vol. 4, tidak. 3, pp. 226–235, 2009. [79] D. Raha, TR Wilson,
acquisition of malignant and stem cell traits,” Nature Reviews J. Peng et al., “The cancer stem cell marker aldehyde dehydrogenase
Cancer, vol. 9, tidak. 4, pp. 265–273, 2009. [68] FA Siebzehnrubl, is required to maintain a drug-tolerant tumor cell subpopulation,”
DJ Silver, B. Tugertimur et al., “The ZEB1 pathway links Cancer Research, vol. 74, tidak. 13, pp. 3579–3590, 2014. [80] DJ
glioblastoma initiation, invasion and chemoresistance,” EMBO Pearce, D. Taussig, C. Simpson et al., “Characterization of cells with
Molecular Medicine, vol. 5, tidak. 8, pp. 1196–1212, 2013. [69] BS a high aldehyde dehydrogenase activity from cord blood and acute
Koo, SH Lee, JM Kim et al., “Oct4 is a critical regu- lator of myeloid leukemia samples,” Stem Cells, vol. 23, tidak. 6, pp. 752–
stemness in head and neck squamous carcinoma cells,” Oncogene, 760, 2005. [81] Y. Su, Q. Qiu, X. Zhang et al., “Aldehyde
vol. 34, tidak. 18, pp. 2317–2324, 2015. [70] CE Huang, CC Yu, dehydrogenase 1 A1–positive cell population is enriched in tumor-
FW Hu, MY Chou, and LL Tsai, “Enhanced chemosensitivity by initiating cells and associated with progression of bladder cancer,”
targeting Nanog in head and neck squamous cell carcinomas,” Can- cer Epidemiology Biomarkers & Prevention, vol. 19, no. 2, pp.
International Journal of Molecular Sciences, vol. 15, tidak. 9, pp. 327–337, 2010. [82] MR Clay, M. Tabor, JH Owen et al., “Single-
14935– 14948, 2014. [71] MY Chou, FW Hu, CH Yu, and CC Yu, marker iden- tification of head and neck squamous cell carcinoma
“Sox2 expres- sion involvement in the oncogenicity and cancer stem cells with aldehyde dehydrogenase,” Head & Neck, vol.
radiochemoresis- tance of oral cancer stem cells,” Oral Oncology, 32, tidak. 9, pp. 1195–1201, 2010. [83] F. Jiang, Q. Qiu, A. Khanna
vol. 51, tidak. 1, pp. 31–39, 2015. [72] GY Chiou, TW Yang, CC et al., “Aldehyde dehydroge- nase 1 is a tumor stem cell-associated
Huang et al., “Musashi-1 promotes a cancer stem cell lineage and marker in lung can- cer,” Molecular Cancer Research, vol. 7, tidak.
chemoresistance in colorectal cancer cells,” Scientific Reports, vol. 3, pp. 330–338, 2009. [84] K. Nakahata, S. Uehara, S. Nishikawa et
7, tidak. 1, hal. 2172, 2017. [73] HZ Cao, XF Liu, WT Yang, Q. al., “Aldehyde dehy- drogenase 1 (ALDH1) is a potential marker for
Chen, and PS Zheng, “LGR5 promotes cancer stem cell traits and cancer stem cells in embryonal rhabdomyosarcoma,” PLoS One, vol.
chemoresistance in cervical cancer,” Cell Death & Disease, vol. 8, 10, tidak. 4, article e0125454, 2015. [85] CP Huang, MF Tsai, TH
tidak. 9, article e3039, 2017. [74] GM Seigel, LM Campbell, M. Chang et al., “ALDH-positive lung cancer stem cells confer
Narayan, and F. Gonzalez- Fernandez, “Cancer stem cell resistance to epidermal growth factor receptor tyrosine kinase
inhibitors,” Cancer Letters, vol. 328, no. 1, pp. 144–151, 2013. [86]cells via an endothelial cell intermediate,” Stem Cells, vol. 28, tidak.
JA Ajani, X. Wang, S. Song et al., “ALDH-1 expression levels 6, pp. 1019–1029, 2010. [103] C. Calabrese, H. Poppleton, M. Kocak
predict response or resistance to preoperative chemora- diation in et al., “A perivascular niche for brain tumor stem cells,” Cancer Cell,
resectable esophageal cancer patients,” Molecular Oncology, vol. 8,vol. 11, no. 1, pp. 69–82, 2007. [104] S. Krishnamurthy, Z. Dong, D.
tidak. 1, pp. 142–149, 2014. [87] A. Kreso, CA O'Brien, P. van Vodopyanov et al., “Endothe- lial cell-initiated signaling promotes the
Galen et al., “Variable clonal repopulation dynamics influence survival and self- renewal of cancer stem cells,” Cancer Research, vol.
chemotherapy response in colorectal cancer,” Science, vol. 339, no.70, no. 23, pp. 9969–9978, 2010. [105] S. Krishnamurthy, KA
6119, pp. 543–548, 2013. [88] A. tumour V. Kurtova, repopulationWarner, Z. Dong et al., “Endothelial interleukin-6 defines the
J. Xiao, abrogates Q. Mo et al., bladder “Blocking cancer tumorigenic potential of primary human cancer stem cells,” Stem
PGEchemoresis- 2-induced Cells, vol. 32, tidak. 11, pp. 2845–2857, 2014. [106] Z. Zhang, Z.
tance,” Nature, vol. 517, no. 7533, pp. 209–213, 2015. [89]Dong, IS Lauxen, MS Filho, and JE Nor, “Endothelial cell-secreted
T. Ishimoto, O. Nagano, T. Yae et al., “CD44 variant regulates redoxEGF induces epithelial to mesen- chymal transition and endows head
status in cancer cells by stabilizing the xCT subunit of system xc − and neck cancer cells with stem-like phenotype,” Cancer Research,
and thereby promotes tumor growth,” Cancer Cell, vol. 19, no. 3, pp.vol. 74, tidak. 10, pp. 2869–2881, 2014. [107] TS Zhu, MA Costello,
387–400, 2011. [90] M. Diehn, RW Cho, NA Lobo et al., CE Talsma et al., “Endothelial cells create a stem cell niche in
“Association of reactive oxygen species levels and radioresistance inglioblastoma by providing NOTCH ligands that nurture self-renewal
can- cer stem cells,” Nature, vol. 458, no. 7239, pp. 780–783, 2009. of cancer stem- like cells,” Cancer Research, vol. 71, no. 18, pp.
[91] H. Yin and J. Glass, “The phenotypic radiation resistance of 6061–6072, 2011. [108] J. Lu, X. Ye, F. Fan et al., “Endothelial cells
CD44+/CD24−or low breast cancer cells is mediated through the promote the colo- rectal cancer stem cell phenotype through a soluble
enhanced activation of ATM signaling,” PLoS One, vol. 6, tidak. 9, form of Jagged-1,” Cancer Cell, vol. 23, tidak. 2, pp. 171–185, 2013.
article e24080, 2011. [92] S. Bao, Q. Wu, RE McLendon et al., [109] S. Schwitalla, AA Fingerle, P. Cammareri et al., “Intestinal
“Glioma stem cells pro- mote radioresistance by preferential tumorigenesis initiated by dedifferentiation and acquisition of stem-
activation of the DNA damage response,” Nature, vol. 444, no. 7120, cell-like properties,” Cell, vol. 152, no. 1-2, pp. 25– 38, 2013. [110]
pp. 756–760, 2006. [93] J. Wang, TP Wakeman, JD Lathia et al., L. Vermeulen, F. de Sousa E Melo, M. van der Heijden et al., “Wnt
“Notch promotes radioresistance of glioma stem cells,” Stem Cells, activity defines colon cancer stem cells and is regulated by the
vol. 28, tidak. 1, pp. 17–28, 2010. microenvironment,” Nature Cell Biology, vol. 12, tidak. 5, pp. 468–
[94] K. Kise, Y. Kinugasa-Katayama, and N. Takakura, “Tumor 476, 2010. [111] XC He, J. Zhang, WG Tong et al., “BMP signaling
microenvironment for cancer stem cells,” Advanced Drug Delivery inhibits intestinal stem cell self-renewal through suppression of Wnt–
Reviews, vol. 99, Part B, pp. 197–205, 2016. [95] EY-T. Lau, NP-Y. β-catenin signaling,” Nature Genetics, vol. 36, tidak. 10, pp. 1117–
Ho, and TK-W. Lee, “Cancer stem cells and their microenvironment: 1121, 2004. [112] LA Milner and A. Bigas, “Notch as a mediator of
biology and therapeutic implications,” Stem Cells International, vol. cell fate determination in hematopoiesis: evidence and speculation,”
2017, Article ID 3714190, 11 pages, 2017. [96] A. Turdo, M. Todaro, Blood, vol. 93, tidak. 8, pp. 2431–2448, 1999. [113] O.-J. Kwon, JM
and G. StassiS. Babashah, “Targeting cancer stem cells and the tumor Valdez, L. Zhang et al., “Increased Notch signalling inhibits anoikis
microenvironment,” in Can- cer Stem Cells: Emerging Concepts and and stimulates proliferation of prostate luminal epithelial cells,”
Future Perspectives in Translational Oncology, pp. 445–476, Springer Nature Communications, vol. 5, p. 4416, 2014. [114] BRB Pires, Í.
International Publishing, Cham, 2015. [97] H. Adisetiyo, M. Liang, SS De Amorim, LDE Souza, JA Rodri- gues, and AL Mencalha,
CP Liao et al., “Dependence of castration-resistant prostate cancer “Targeting cellular signaling path- ways in breast cancer stem cells
(CRPC) stem cells on CRPC-associated fibroblasts,” Journal of and its implication for cancer treatment,” Anticancer Research, vol.
Cellular Physiology, vol. 229, no. 9, pp. 1170–1176, 2014. [98] CP 36, tidak. 11, pp. 5681– 5692, 2016. [115] B. Bao, AS Azmi, S. Ali et
Liao, H. Adisetiyo, M. Liang, and P. Roy-Burman, “Can- cer- al., “The biological kinship of hyp- oxia with CSC and EMT and their
associated fibroblasts enhance the gland-forming capabil- ity of relationship with deregu- lated expression of miRNAs and tumor
prostate cancer stem cells,” Cancer Research, vol. 70, no. 18, pp. aggressiveness,” Biochimica et Biophysica Acta (BBA) - Reviews on
7294–7303, 2010. [99] W.-J. Chen, C.-C. Ho, Y.-L. Chang et al., Cancer, vol. 1826, no. 2, pp. 272–296, 2012. [116] B. Bao, S. Ali, A.
“Cancer-associated fibroblasts regulate the plasticity of lung cancer Ahmad et al., “Hypoxia-induced aggressive- ness of pancreatic cancer
stemness via paracrine signalling,” Nature Communications, vol. 5, p. cells is due to increased expression of VEGF, IL-6 and miR-21,
3472, 2014. [100] Y. Kinugasa, T. Matsui, and N. Takakura, “CD44 which can be attenuated by CDF treatment,” PLoS One, vol. 7, tidak.
expressed on cancer-associated fibroblasts is a functional molecule 12, article e50165, 2012. [117] KP Ng, A. Manjeri, KL Lee et al.,
sup- porting the stemness and drug resistance of malignant cancer “Physiologic hypoxia promotes maintenance of CML stem cells
cells in the tumor microenvironment,” Stem Cells, vol. 32, tidak. 1, despite effective BCR-ABL1 inhibition,” Blood, vol. 123, no. 21, pp.
pp. 145–156, 2014. [101] F. Cammarota and MO Laukkanen, 3316– 3326, 2014. [118] A. Murakami, F. Takahashi, F. Nurwidya et
“Mesenchymal stem/ stromal cells in stromal evolution and cancer al., “Hypoxia increases gefitinib-resistant lung cancer stem cells
progression,” Stem Cells International, vol. 2016, Article ID 4824573, through the activation of insulin-like growth factor 1 receptor,” PLoS
11 pages, 2016. [102] KE Hovinga, F. Shimizu, R. Wang et al., One, vol. 9, tidak. 1, article e86459, 2014. [119] V. Rausch, L. Liu,
“Inhibition of notch signaling in glioblastoma targets cancer stem A. Apel et al., “Autophagy mediates sur- vival of pancreatic tumour-
initiating cells in a hypoxic micro- environment,” The Journal of 2017. [122] P. Luraghi, G. Reato, E. Cipriano et al., “MET signaling
Pathology, vol. 227, no. 3, pp. 325–335, 2012. [120] F. Lotti, AM in colon cancer stem-like cells blunts the therapeutic response to
Jarrar, RK Pai et al., “Chemotherapy activates cancer-associated EGFR inhibitors,” Cancer Research, vol. 74, tidak. 6, pp. 1857–1869,
fibroblasts to maintain colorectal cancer- initiating cells by IL-17A,” 2014. [123] H. Korkaya, Gi Kim, A. Davis et al., “Activation of an
Journal of Experimental Medicine, vol. 210, no. 13, pp. 2851–2872, IL6 inflammatory loop mediates trastuzumab resistance in HER2+
2013. [121] N. Nair, AS Calle, MH Zahra et al., “A cancer stem cell breast cancer by expanding the cancer stem cell pop- ulation,”
model as the point of origin of cancer-associated fibroblasts in tumor Molecular Cell, vol. 47, tidak. 4, pp. 570–584, 2012.
microenvironment,” Scientific Reports, vol. 7, tidak. 1, hal. 6838,
12 Stem Cells International
[124] NE Bhola, JM Balko, TC Dugger et al., “TGF-β inhibi- tion melanoma that pro- motes metastasis and chemoresistance,” Genes &
enhances chemotherapy action against triple-negative breast cancer,” Develop- ment, vol. 30, no. 1, pp. 18–33, 2016. [138] E. Proctor, M.
The Journal of Clinical Investigation, vol. 123, no. 3, pp. 1348–1358, Waghray, CJ Lee et al., “Bmi1 enhances tumorigenicity and cancer
2013. [125] YG Yang, IN Sari, MF Zia, SR Lee, SJ Song, and HY stem cell function in pancreatic adenocarcinoma,” PLoS One, vol. 8,
Kwon, “Tetraspanins: spanning from solid tumors to hema- tologic tidak. 2, article e55820, 2013.
malignancies,” Experimental Hematology, vol. 44, tidak. 5, pp. 322– [139] E. Wang, S. Bhattacharyya, A. Szabolcs et al., “Enhancing
328, 2016. [126] HY Kwon, J. Bajaj, T. Ito et al., “Tetraspanin 3 is chemotherapy response with Bmi-1 silencing in ovarian can- cer,”
required for the development and propagation of acute myelogenous PLoS One, vol. 6, tidak. 3, article e17918, 2011. [140] J. Wu, D. Hu,
leukemia,” Cell Stem Cell, vol. 17, tidak. 2, pp. 152–164, 2015. [127] and R. Zhang, “Depletion of Bmi-1 enhances 5- fluorouracil-induced
Z. Zeng, IJ Samudio, M. Munsell et al., “Inhibition of CXCR4 with apoptosis and autophagy in hepatocel- lular carcinoma cells,”
the novel RCP168 peptide overcomes stroma- mediated Oncology Letters, vol. 4, tidak. 4, pp. 723–726, 2012. [141] AV
chemoresistance in chronic and acute leukemias,” Molecular Cancer Ougolkov, VN Bilim, and DD Billadeau, “Regulation of pancreatic
Therapeutics, vol. 5, tidak. 12, pp. 3113– 3121, 2006. [128] AK tumor cell proliferation and chemoresistance by the histone
Azab, JM Runnels, C. Pitsillides et al., “CXCR4 inhib- itor AMD3100 methyltransferase enhancer of zeste homologue 2,” Clinical Cancer
disrupts the interaction of multiple myeloma cells with the bone Research, vol. 14, tidak. 21, pp. 6790–6796, 2008. [142] SH Kim, K.
marrow microenvironment and enhances their sensitivity to therapy,” Joshi, R. Ezhilarasan et al., “EZH2 protects gli- oma stem cells from
Blood, vol. 113, no. 18, pp. 4341– 4351, 2009. [129] T. Yamashina, radiation-induced cell death in a MELK/ FOXM1-dependent
M. Baghdadi, A. Yoneda et al., “Cancer stem- like cells derived from manner,” Stem Cell Reports, vol. 4, tidak. 2, pp. 226–238, 2015.
chemoresistant tumors have a unique capacity to prime tumorigenic [143] CM Fillmore, C. Xu, PT Desai et al., “EZH2 inhibition
myeloid cells,” Cancer Research, vol. 74, tidak. 10, pp. 2698–2709, sensitizes BRG1 and EGFR mutant lung tumours to TopoII
2014. [130] Y. Zheng, Z. Cai, S. Wang et al., “Macrophages are an inhibitors,” Nature, vol. 520, no. 7546, pp. 239–242, 2015. [144] AM
abun- dant component of myeloma microenvironment and protect Pietersen, HM Horlings, M. Hauptmann et al., “EZH2 and BMI1
myeloma cells from chemotherapy drug–induced apoptosis,” Blood, inversely correlate with prognosis and TP53 mutation in breast
vol. 114, no. 17, pp. 3625–3628, 2009. [131] JB Mitchem, DJ cancer,” Breast Cancer Research, vol. 10, tidak. 6, article R109, 2008.
Brennan, BL Knolhoff et al., “Targeting tumor-infiltrating [145] B. Zhang, AC Strauss, S. Chu et al., “Effective targeting of
macrophages decreases tumor-initiating cells, relieves quiescent chronic myelogenous leukemia stem cells by histone
immunosuppression, and improves chemother- apeutic responses,” deacetylase inhibitors in combination with ima- tinib mesylate,”
Cancer Research, vol. 73, tidak. 3, pp. 1128– 1141, 2013. [132] M. Cancer Cell, vol. 17, tidak. 5, pp. 427–442, 2010. [146] FM Frame,
Amit and Z. Gil, “Macrophages increase the resistance of pancreatic D. Pellacani, AT Collins et al., “HDAC inhib- itor confers
adenocarcinoma cells to gemcitabine by upregu- lating cytidine radiosensitivity to prostate stem-like cells,” Brit- ish Journal of
deaminase,” Oncoimmunology, vol. 2, tidak. 12, article e27231, 2013. Cancer, vol. 109, tidak. 12, pp. 3023–3033, 2013. [147] F. Bruzzese,
[133] OG McDonald, H. Wu, W. Timp, A. Doi, and AP Fein- berg, A. Leone, M. Rocco et al., “HDAC inhibitor vor- inostat enhances the
“Genome-scale epigenetic reprogramming during epithelial-to- antitumor effect of gefitinib in squa- mous cell carcinoma of head and
mesenchymal transition,” Nature Structural & Molecular Biology, neck by modulating ErbB receptor expression and reverting EMT,”
vol. 18, tidak. 8, pp. 867–874, 2011. [134] WJ Harris, X. Huang, JT Journal of Cellular Physiology, vol. 226, tidak. 9, pp. 2378–2390,
Lynch et al., “The histone demethylase KDM1A sustains the 2011. [148] K.-H. Song, CH Choi, H.-J. Lee et al., “HDAC1
oncogenic potential of MLL-AF9 leukemia stem cells,” Cancer Cell, upregula- tion by NANOG promotes multidrug resistance and a stem-
vol. 21, tidak. 4, pp. 473–487, 2012. [135] F. Crea, R. Danesi, and like phenotype in immune edited tumor cells,” Cancer Research, vol.
WL Farrar, “Cancer stem cell epige- netics and chemoresistance,” 77, no. 18, 2017. [149] I. Ibanez de Caceres, M. Cortes-Sempere, C.
Epigenomics, vol. 1, tidak. 1, pp. 63–79, 2009. [136] L. Qin, X. Moratilla et al., “IGFBP-3 hypermethylation-derived deficiency
Zhang, L. Zhang et al., “Downregulation of BMI-1 enhances 5- mediates cis- platin resistance in non-small-cell lung cancer,”
fluorouracil-induced apoptosis in nasopharyn- geal carcinoma cells,” Oncogene, vol. 29, tidak. 11, pp. 1681–1690, 2010. [150] G.
Biochemical and Biophysical Research Communications, vol. 371, Strathdee, MJ MacKean, M. Illand, and R. Brown, “A role for
no. 3, pp. 531–535, 2008. [137] R. Ferretti, A. Bhutkar, MC methylation of the hMLH1 promoter in loss of hMLH1 expression
McNamara, and JA Lees, “BMI1 induces an invasive signature in and drug resistance in ovarian cancer,” Oncogene, vol. 18, tidak. 14,
pp. 2335–2341, 1999. [151] G. Gifford, J. Paul, PA Vasey, SB Kaye, treatment with cytotoxic anticancer agents,” Oncogene, vol. 15, tidak.
and R. Brown, “The acquisition of hMLH1 methylation in plasma 1, pp. 45–52, 1997. [153] HJ Mackay, D. Cameron, M. Rahilly et al.,
DNA after chemotherapy predicts poor survival for ovarian cancer “Reduced MLH1 expression in breast tumors after primary
patients,” Clinical Cancer Research, vol. 10, tidak. 13, pp. 4420– chemotherapy pre- dicts disease-free survival,” Journal of Clinical
4426, 2004. [152] R. Brown, GL Hirst, WM Gallagher et al., Oncology, vol. 18, tidak. 1, pp. 87–93, 2000.
“hMLH1 expression and cellular responses of ovarian tumour cells to
13 Stem Cells International
[154] JP Sullivan, M. Spinola, M. Dodge et al., “Aldehyde dehy- Molecular Medicine, vol. 17, tidak. 1-2, pp. 103–112, 2011. [168] RD
drogenase activity selects for lung adenocarcinoma stem cells Meng, CC Shelton, YM Li et al., “γ-Secretase inhib- itors abrogate
dependent on notch signaling,” Cancer Research, vol. 70, no. 23, pp. oxaliplatin-induced activation of the Notch-1
9937–9948, 2010. [155] G. Dontu, KW Jackson, E. McNicholas, MJ signaling pathway in colon cancer cells resulting in enhanced
Kawamura, WM Abdallah, and MS Wicha, “Role of Notch signaling chemosensitivity,” Cancer Research, vol. 69, tidak. 2, pp. 573– 582,
in cell-fate determination of human mammary stem/progen- itor 2009. [169] SM McAuliffe, SL Morgan, GA Wyant et al., “Targeting
cells,” Breast Cancer Research, vol. 6, tidak. 6, pp. R605– R615, Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors
2004. [156] DM Park, J. Jung, J. Masjkur et al., “Hes3 regulates cell to platinum therapy,” Proceedings of the National Academy of
number in cultures from glioblastoma multiforme with stem cell Sciences of the United States of America, vol. 109, tidak. 43, pp.
characteristics,” Scientific Reports, vol. 3, tidak. 1, hal. 1095, 2013. E2939–E2948, 2012. [170] YP Liu, CJ Yang, MS Huang et al.,
[157] L. Yang, G. Xie, Q. Fan, and J. Xie, “Activation of the “Cisplatin selects for multidrug-resistant CD133+ cells in lung
hedgehog-signaling pathway in human cancer and the clini- cal adenocarcinoma by activating Notch signaling,” Cancer Research,
implications,” Oncogene, vol. 29, tidak. 4, pp. 469–481, 2010. [158] vol. 73, tidak. 1, pp. 406–416, 2013. [171] S. Alonso, RJ Jones, and
WF Tam, PS Hahnel, A. Schuler et al., “STAT5 is crucial to maintain G. Ghiaur, “Retinoic acid, CYP26, and drug resistance in the stem cell
leukemic stem cells in acute myelogenous leuke- mias induced by niche,” Experimental Hematology, vol. 54, pp. 17–25, 2017. [172] M.
MOZ-TIF2,” Cancer Research, vol. 73, tidak. 1, pp. 373–384, 2013. Krause, A. Dubrovska, A. Linge, and M. Baumann, “Can- cer stem
[159] N. Takebe, L. Miele, PJ Harris et al., “Targeting Notch, cells: radioresistance, prediction of radiotherapy outcome and specific
Hedgehog, and Wnt pathways in cancer stem cells: clinical update,” targets for combined treatments,” Advanced Drug Delivery Reviews,
Nature Reviews Clinical Oncology, vol. 12, tidak. 8, pp. 445–464, vol. 109, pp. 63–73, 2017. [173] R. Olson and C. Albright, “Recent
2015. [160] I. Kurth, C. Peitzsch, M. Baumann, and A. Dubrovska, progress in the medicinal chemistry of γ-secretase inhibitors,” Current
“The role of cancer stem cells in tumor Radioresistance,” in Cancer Topics in Medic- inal Chemistry, vol. 8, tidak. 1, pp. 17–33, 2008.
Stem Cells, pp. 473–491, John Wiley & Sons, Hoboken, New Jersey, [174] S. Richter, PL Bedard, EX Chen et al., “A phase I study of the
USA, 2014. [161] T. Noda, H. Nagano, I. Takemasa et al., oral gamma secretase inhibitor R04929097 in combina- tion with
“Activation of Wnt/ β-catenin signalling pathway induces gemcitabine in patients with advanced solid tumors (PHL-078/CTEP
chemoresistance to interferon-α/5-fluorouracil combination therapy 8575),” Investigational New Drugs, vol. 32, tidak. 2, pp. 243–249,
for hepato- cellular carcinoma,” British Journal of Cancer, vol. 100, 2014. [175] PA Beachy, SG Hymowitz, RA Lazarus, DJ Leahy, and
no. 10, pp. 1647–1658, 2009. [162] W. Yang, HX Yan, L. Chen et al., C. Siebold, “Interactions between Hedgehog proteins and their
“Wnt/β-catenin signaling contributes to activation of normal and binding partners come into view,” Genes & Develop- ment, vol. 24,
tumorigenic liver progenitor cells,” Cancer Research, vol. 68, tidak. tidak. 18, pp. 2001–2012, 2010. [176] L. Dirix, “Discovery and
11, pp. 4287– 4295, 2008. [163] M. Flahaut, R. Meier, A. Coulon et exploitation of novel targets by approved drugs,” Journal of Clinical
al., “The Wnt receptor FZD1 mediates chemoresistance in Oncology, vol. 32, tidak. 8, pp. 720-721, 2014. [177] A. Sekulic, MR
neuroblastoma through activation of the Wnt/β-catenin pathway,” Migden, AE Oro et al., “Efficacy and safety of vismodegib in
Oncogene, vol. 28, tidak. 23, pp. 2245–2256, 2009. [164] WK Chau, advanced basal-cell carcinoma,” The New England Journal of
CK Ip, ASC Mak, HC Lai, and AST Wong, “c-Kit mediates Medicine, vol. 366, no. 23, pp. 2171– 2179, 2012. [178] X. Li, V.
chemoresistance and tumor- initiating capacity of ovarian cancer cells Placencio, JM Iturregui et al., “Prostate tumor pro- gression is
through activation of Wnt/β-catenin–ATP-binding cassette G2 mediated by a paracrine TGF-β/Wnt3a signaling axis,” Oncogene,
signaling,” Oncogene, vol. 32, tidak. 22, pp. 2767–2781, 2013. [165] vol. 27, tidak. 56, pp. 7118–7130, 2008. [179] M. Kahn, “Can we
X. Zeng, H. Zhao, Y. Li et al., “Targeting Hedgehog signaling safely target the WNT pathway?,” Nature
pathway and autophagy overcomes drug resistance of BCR- ABL- Reviews Drug Discovery, vol. 13, no. 7, pp. 513–532, 2014. [180]
positive chronic myeloid leukemia,” Autophagy, vol. 11, no. 2, pp. DC Smith, LS Rosen, R. Chugh et al., “First-in-human evaluation of
355–372, 2015. [166] M. Xu, A. Gong, H. Yang et al., “Sonic the human monoclonal antibody vantictumab (OMP-18R5; anti-
hedgehog-glioma asso- ciated oncogene homolog 1 signaling Frizzled) targeting the WNT pathway in a phase I study for patients
enhances drug resis- tance in CD44+/Musashi-1+ gastric cancer stem with advanced solid tumors,” Jour- nal of Clinical Oncology, vol. 31,
cells,” Cancer Letters, vol. 369, no. 1, pp. 124–133, 2015. [167] IV 15 Supplement, p. 2540, 2013. [181] US National Library of
Ulasov, S. Nandi, M. Dey, AM Sonabend, and MS Lesniak, Medicine, “ClinicalTrials.Gov,”
“Inhibition of Sonic hedgehog and Notch pathways enhances 2014, http://clinicaltrials.gov/show/NCT01345201. [182]
sensitivity of CD133+ glioma stem cells to temozo- lomide therapy,” A. Jimeno, MS Gordon, R. Chugh et al., “A first-in-human phase 1
study of anticancer stem cell agent OMP-54F28 (FZD8-Fc), decoy and reprogramming: origins of cancer stem cells,” EMBO Reports,
receptor for WNT ligands, in patients with advanced solid tumors,” vol. 15, tidak. 3, pp. 244–253, 2014. [184] B. Du and J. Shim,
Journal of Clinical Oncology, vol. 32, 15 Supplement, p. 2505, 2014. “Targeting epithelial–mesenchymal transition (EMT) to overcome
[183] D. Friedmann-Morvinski and IM Verma, “Dedifferentia- tion drug resistance in cancer,” Molecules, vol. 21, tidak. 8, 2016.
14 Stem Cells International
Stem Cells International 15
[200] [185] R. Malek, H. Wang, K. Taparra, and PT Tran, “Therapeutic P. Bu, KY Chen, JH Chen et al., “A m
targeting of epithelial plasticity pro
regulated bimodal switch targets Notch in colon cancer stem epithelial-mesenchymal transition,” Cells, Tissues, Organs,
cells,” Cell Stem Cell, vol. 12, tidak. 5, pp. 602–615, 2013. vol. 203, no. 2, pp. 114–127, 2017.
[201] C. Liu, K. Kelnar, B. Liu et al., “The microRNA miR-34a [186] S. Knoll, S. Emmrich, and BM Putzer, “The E2F1-miRNA
inhibits prostate cancer stem cells and metastasis by directly cancer progression network,” Advances in Experimental
repressing CD44,” Nature Medicine, vol. 17, tidak. 2, pp. 211– Medicine and Biology, vol. 774, pp. 135–147, 2013.
215, 2011. [187] AA Dar, S. Majid, D. de Semir, M. Nosrati, V. Bezrookove,
[202] YY Wu, YL Chen, YC Jao, IS Hsieh, KC Chang, and and M. Kashani-Sabet, “miRNA-205 suppresses melanoma
TM Hong, “miR-320 regulates tumor angiogenesis driven cell proliferation and induces senescence via regulation of
by vascular endothelial cells in oral cancer by silencing neuro- E2F1 protein,” Journal of Biological Chemistry, vol. 286,
pilin 1,” Angiogenesis, vol. 17, tidak. 1, pp. 247–260, 2014. no. 19, pp. 16606–16614, 2011.
[203] C. Visus, Y. Wang, A. Lozano-Leon et al., “Targeting ALDHb- [188] V. Alla, BS Kowtharapu, D. Engelmann et al., “E2F1
right human carcinoma–initiating cells with ALDH1A1- confers anticancer drug resistance by targeting ABC trans-
specific CD8+ T cells,” Clinical Cancer Research, vol. 17, porter family members and Bcl-2 via the p73/DNp73-miR-
no. 19, pp. 6174–6184, 2011. 205 circuitry,” Cell Cycle, vol. 11, no. 16, pp. 3067–3078, 2012.
[204] L. Jin, KJ Hope, Q. Zhai, F. Smadja-Joffe, and JE Dick, “Targeting of CD44 eradicates human acute myeloid leuke- [189] PA Gregory, AG B

mic stem cells,” Nature Medicine, vol. 12, tidak. 10, pp. 1167– family and miR-205 regulate epithelial to mesenchymal tran-
1174, 2006. sition by targeting ZEB1 and SIP1,” Nature Cell Biology, vol. 10, tidak. 5, pp. 593–601, 2008.
[205] Y. Guo, J. Ma, J. Wang et al., “Inhibition of human melanoma growth and metastasis in vivoby anti-CD44 monoclonal anti- [190] C. Palena
“Immun
body,” Cancer Research, vol. 54, tidak. 6, pp. 1561–1565, 1994. epithelial–mesenchymal transition via brachyury-based vac- cines,” Advances in Can
128, pp. 69–93, 2015.
[206] SK Swaminathan, MR Olin, CL Forster, KSS Cruz, J. Panyam, and JR Ohlfest, “Identification of a novel mono- clonal antibody recognizing
Immunolog- [191] J. Godlewski, MO Nowicki, A. Bronisz
ical Methods, vol. 361, no. 1-2, pp. 110–115, 2010. the Bmi-1 oncogene/stem cell renewal factor by microRNA- 128 inhibits glioma proliferation and
Cancer Research, vol. 68, tidak. 22, pp. 9125–9130, 2008.
[207] F. Ferrari, S. Bellone, J. Black et al., “Solitomab, an EpCAM/ CD3 bispecific antibody construct (BiTE®), is highly active against primary
carcinosarcoma cell lines [192] CA O'Brien, A. Kreso, P. Ryan et al., “ID
in vitro,” Journal of Experimental & Clinical Cancer Research, the self-renewal capacity of human colon cancer-initiating
vol. 34, tidak. 1, hal. 123, 2015. cells through p21,” Cancer Cell, vol. 21, tidak. 6, pp. 777–792, 2012.
[208] M. Sen, DM Wankowski, NK Garlie et al., “Use of anti- CD3 × anti-HER2/neu bispecific antibody for redirecting [193] J. Deng, M. Yang,
Wang
cytotoxicity of activated T cells toward HER2/neu+ tumors,” non-coding RNA HOTAIR regulates the proliferation, self-
Journal of Hematotherapy & Stem Cell Research, vol. 10, renewal capacity, tumor formation and migration of the can-
no. 2, pp. 247–260, 2001. cer stem-like cell (CSC) subpopulation enriched from breast cancer cells,” PLoS One, vol. 12, tidak. 1, article e0170860, 20
[209] F. Dammeijer, LA Lievense, ME Kaijen-Lambers et al., “Depletion of tumor-associated macrophages with a CSF- [194] SJ Song, L. Polise

1R kinase inhibitor enhances antitumor immunity and nism regulates breast cancer stemness and metastasis via
survival induced by DC immunotherapy,” Cancer Immunol- TET-family-dependent chromatin remodeling,” Cell,
ogy Research, vol. 5, tidak. 7, pp. 535–546, 2017. vol. 154, no. 2, pp. 311–324, 2013.
[210] DR Yang, XF Ding, J. Luo et al., “Increased chemosensitiv- [195] F. Yu, H. Yao, P. Zhu et al., “let-7 regulates self renewal and
ity via targeting testicular nuclear receptor 4 (TR4)-Oct4- tumorigenicity of breast cancer cells,” Cell, vol. 131, no. 6,
interleukin 1 receptor antagonist (IL1Ra) axis in prostate pp. 1109–1123, 2007.
cancer CD133+ stem/progenitor cells to battle prostate [196] Y. Shimono, M. Zabala, RW Cho et al., “Downregulation of miRNA-200c links breast ca
normal stem
cancer,” Journal of Biological Chemistry, vol. 288, no. 23, pp. 16476–16483, 2013. cells,” Cell, vol. 138, no. 3,
[211] SY Kim, JW Kang, X. Song et al., “Role of the IL-6-JAK1- [197] D. Iliopoulos, M. Lindahl-Allen, C. Polytarchou, HA Hirsch, PN Tsichlis, and
miR-200 inhibi- tion of Suz12 leads to polycomb-mediated repression
STAT3-Oct-4 pathway in the conversion of non-stem cancer cells into cancer stem-like cells,” Cellular Signalling, vol. 25, tidak. 4, pp. 961–969, 20
formation and mainten
[212] C. Ginestier, S. Liu, ME Diebel et al., “CXCR1 blockade cells,” Molecular Cell, vol. 39, no. 5, pp. 761–772, 2010.
selectively targets human breast cancer stem cells in vitro [198] D. Iliopoulos, A. Rotem, and K. Struhl, “Inhibition of miR- 193a expression by Max a
K-Ras and
and in xenografts,” The Journal of Clinical Investigation, vol. 120, no. 2, pp. 485–497, 2010. PLAU to mediate distinct aspects of cellu
[213] SL Topalian, FS Hodi, JR Brahmer et al., “Safety, activity, Cancer Research, vol. 71, no. 15, pp. 5144–5153, 2011.
and immune correlates of anti–PD-1 antibody in cancer,” [199] N. Bitarte, E. Bandres, V. Boni et al., “MicroRNA-451 is involved in the self-renewal,
and chemoresis-
The New England Journal of Medicine, vol. 366, no. 26, pp. 2443–2454, 2012. tance of colorectal cancer stem cells,”
[214] SM Ansell, AM Lesokhin, I. Borrello et al., “PD-1 blockade no. 11, pp. 1661–1671, 2011.
with nivolumab in relapsed or refractory Hodgkin's
16 Stem Cells International

lymphoma,” The New England Journal of Medicine, vol. 372, no. 4, pp. 311–319, 2015. [215] Y. Hu and L.
Fu, “Targeting cancer stem cells: a new therapy to cure cancer patients,” American Journal of Cancer
Research, vol. 2, tidak. 3, pp. 340–356, 2012. [216] SR Pine, B. Marshall, and L. Varticovski, “Lung cancer
stem
cells,” Disease Markers, vol. 24, tidak. 4-5, pp. 257–266, 2008. [217] T. Chanmee, P. Ontong, K.
Kimata, and N. Itano, “Key roles of hyaluronan and its CD44 receptor in the stemness and sur- vival of cancer
stem cells,” Frontiers in Oncology, vol. 5, 2015. [218] YJ Kim, EL Siegler, N. Siriwon, and P. Wang,
“Therapeutic strategies for targeting cancer stem cells,” Journal of Cancer Metastasis and Treatment, vol. 2,
tidak. 7, pp. 233–242, 2016. [219] DL Dragu, LG Necula, C. Bleotu, CC Diaconu, and M. Chivu-
Economescu, “Therapies targeting cancer stem cells: current trends and future challenges,” World Journal of
Stem Cells, vol. 7, tidak. 9, pp. 1185–1201, 2015. [220] L. Han, S. Shi, T. Gong, Z. Zhang, and X. Sun,
“Cancer stem cells: therapeutic implications and perspectives in cancer therapy,” Acta Pharmaceutica Sinica
B, vol. 3, tidak. 2, pp. 65–75, 2013. [221] C. Zhang, C. Li, F. He, Y. Cai, and H. Yang, “Identification of
CD44+CD24+ gastric cancer stem cells,” Journal of Cancer Research and Clinical Oncology, vol. 137, no. 11,
pp. 1679–1686, 2011. [222] S. Takaishi, T. Okumura, S. Tu et al., “Identification of gastric cancer stem cells
using the cell surface marker CD44,” Stem Cells, vol. 27, tidak. 5, pp. 1006–1020, 2009. [223] AT Collins,
PA Berry, C. Hyde, MJ Stower, and NJ Maitland, “Prospective identification of tumorigenic prostate cancer
stem cells,” Cancer Research, vol. 65, no. 23, pp. 10946–10951, 2005. [224] MJ Son, K. Woolard, DH Nam,
J. Lee, and HA Fine, “SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma,” Cell
Stem Cell, vol. 4, tidak. 5, pp. 440–452, 2009. [225] ME Prince, R. Sivanandan, A. Kaczorowski et al.,
“Identifi- cation of a subpopulation of cells with cancer stem cell prop- erties in head and neck squamous cell
carcinoma,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104,
tidak. 3, pp. 973–978, 2007. [226] SK Singh, C. Hawkins, ID Clarke et al., “Identification of human brain
tumour initiating cells,” Nature, vol. 432, no. 7015, pp. 396–401, 2004.
Bioinformatics
Advances in
Hindawi www.hindawi.com Volume 2018

Enzyme Research
Hindawi www.hindawi.com Volume 2018

Anatomy Research International


Hindawi www.hindawi.com Volume 2018

The World Scientific Journal Hindawi Hindawi

Publishing Corporation http://www.hindawi.com www.hindawi.com


Volume Volume 2018 2013

International Genomics
Journal of
Hindawi www.hindawi.com Volume 2018
Advances in Virology
Hindawi www.hindawi.com Volume 2018

Journal of Parasitology Research


Hindawi www.hindawi.com Volume 2018

Submit your manuscripts at www.hindawi.com


International
Hindawi www.hindawi.com Volume 2018
Peptides Journal of

Neuroscience Journal
Hindawi www.hindawi.com Volume 2018

Zoology
International

Journal of
Hindawi www.hindawi.com
Volume 2018

Marine of
Journal

Biology
Hindawi www.hindawi.com Volume 2018

Stem Cells International


Hindawi www.hindawi.com Volume 2018

International Journal of Microbiology


Hindawi www.hindawi.com Volume 2018

Biochemistry Research Inte rnational


Hindawi www.hindawi.com Volume 2018

BioMed Research International


Hindawi www.hindawi.com Volume 2018

Genetics Research International


Hindawi www.hindawi.com Volume 2018

Archaea
Hindawi www.hindawi.com Volume 2018

Anda mungkin juga menyukai