Anda di halaman 1dari 17

BBA - Ulasan tentang Cancer 1874 (2020) 188410

Daftar isi tersedia di ScienceDirect

BBA - Ulasan tentangCancer


beranda jurnal: www.elsevier.com/locate/bbacan

Review

Human MYC G-quadruplex: Dari penemuan hingga target terapi kanker T


a,⁎
Wenmeng Wanga, Shuangli Hua, Yaru Gua, Yunxiao Yana, Daniel B.Stovallb, Dangdang Li ,
a,
Guangchao Sui ⁎

a b
Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
College of Arts and Sciences, Winthrop University, Rock Hill, SC 29733, Amerika Serikat

INFO ARTIKEL dan metastasis, yang menyebabkan hasil klinis yang buruk [8]. Karena
perannya dalam
Kata kunci: mengatur transkripsi global [11], downregulasi MYC adalah mantan
MYC Kanker adalah penyakit utama yang menimbulkan ancaman serius bagi
Ekspresi gen kesehatan manusia di seluruh dunia, terutama di negara berkembang [1,2].
G-quadruplex (G4)
Agen pengikat G4
Pada tingkat molekuler, kanker adalah kelainan genetik dan kejadiannya
peptida terkait erat dengan ekspresi menyimpang dari penekan tumor dan onkogen
terapi kanker [3,4]. Di antara onkogen ini, MYC diekspresikan secara berlebihan hingga
70% dari semua kanker pada manusia, dan terkait dengan sekitar 70.000
kematian per tahun di Amerika Serikat [5–7]. Produk gen MYC adalah
faktor transkripsi utama yang mengikat E-box tertentu di promotor target
saat membentuk heterodimer dengan MAX dan mengontrol ekspresi
banyak gen target, penting untuk banyak kemajuan fisiologis, seperti
proliferasi sel, diferensiasi, adhesi , apoptosis, angio genesis dan
metastasis [6,8-10]. Dengan demikian, peningkatan level MYC secara
langsung berkontribusi pada ekspresi gen yang berubah yang mendorong
1. Pendahuluan perkembangan kanker yang
ABSTRAK diharapkan dapat mengurangi proliferasi dan kelangsungan hidup semua
jenis sel. Namun, penghambatan transkripsi MYC menyebabkan hilangnya
Ekspresi berlebihan dari onkogen MYC adalah ciri molekuler dari inisiasi dan fenotipe ganas secara dramatis di sebagian besar sel tumor dengan sedikit
perkembangan kanker. Target MYC adalah strategi terapi kanker yang logis dan atau tanpa toksisitas pada sel normal [12-15]. Dengan demikian,
efektif. Struktur sekunder DNA khusus, G quadruplex (G4), terbentuk di dalam penargetan MYC menjanjikan sebagai strategi yang efisien dan aman
wilayah nuclease hipersensitivitas elemen III 1 (NHE III1), terletak di hulu promotor dalam pengobatan kanker pada manusia.
P1 gen MYC yang menggerakkan sebagian besar transkripsi. Menargetkan struktur Ekspresi MYC yang menyimpang pada kanker terutama diatur pada
G4 seperti itu telah menjadi fokus terapi antikanker dalam beberapa dekade tingkat transkripsi [16]. Unsur hipersensitivitas nuklease III 1 (NHE III1),
terakhir. Dengan demikian, tinjauan komprehensif terhadap struktur MYC G4 dan terletak di hulu promotor P1 MYC, berkontribusi pada 80-90% transkripsi
perannya sebagai target terapi potensial tepat waktu. Dalam ulasan ini, pertama- MYC [16-18]. Khususnya, elemen NHE III1 mengandung daerah kaya G
tama kami menguraikan penemuan struktur MYC G4 dan bukti pembentukannya yang dapat dilipat menjadi struktur sekunder DNA tertentu yang dikenal
secara in vitro dan dalam sel. Kemudian, kami menjelaskan peran fungsional G4 sebagai G-quadruplex (G4), yang secara negatif mengatur transkripsi
dalam mengatur ekspresi gen MYC. Kami juga merangkum tiga jenis protein yang [19,20]. Sebagai salah satu struktur DNA non-kanonik, G4 umumnya terdiri
berinteraksi dengan MYC G4 yang dapat mempromosikan, menstabilkan, dan dari tiga atau lebih G-quartet (juga dikenal sebagai G
melepaskan struktur G4. Akhirnya, kami membahas molekul pengikat G4 dan
aktivitas antikanker dari ligan penstabil G4, termasuk senyawa molekuler kecil dan
peptida, dan menilai potensinya sebagai terapi antikanker baru.

Singkatan: ADAR1, Adenosine deaminase yang bekerja pada RNA 1; AML, Acute myelocytic leukemia; BCL2, B cell leukemia / lymphoma 2 ; BLM, protein sindrom
Bloom; CD, Circular dichroism; ChIP, Chromatin immunoprecipitation; CLSM, Mikroskopi pemindaian laser confocal; CNBP, protein pengikat asam nukleat jari seng tipe
CCHC; DDX5, Asp-Glu-Ala-Asp (DEAD) -460 kotak polipeptida 5; DMS, Dimetilsulfat; EMSA, Uji pergeseran mobilitas elektroforetik; FRET, transfer energi resonansi
fluoresensi; FUSE, elemen urutan hulu jauh; G4, G-quadruplex; HIF-1α, Faktor yang diinduksi hipoksia 1-alfa; hnRNP K, Ribonukleoprotein nuklir heterogen K; hPARP-
1, poli (ADP-ribosa) polimerase-1; ITC, kalorimetri titrasi isotermal; NAD, Nicotinamide adenine dinucleotide; NCL, Nucleolin; NF Y, Nuclear factor Y; NHE III 1, Nuclease
hipersensitivitas elemen III1; NMR, Resonansi magnetik nuklir; NPM1, Nukleofos min; PDGF-A, faktor pertumbuhan A yang diturunkan dari trombosit; RBD, domain
pengikat RNA; RECQ5, RecQ seperti helicase 5; RGG, Arginin-glisin-glisin; SP1, Kekhususan protein 1; SPR, Resonansi permukaan plasmon; TNBC, kanker payudara
triple-negatif; TSS, Situs inisiasi Transkripsi; WRN, protein sindrom Werner; XRCC1, perbaikan silang sinar-X melengkapi 1; ε-PLL, ε-poly-l-lysine; α, ε-PLL, α, ε-poly-l-
lysine; 2-Ap, 2-aminopurine

Sesuai penulis.
Alamat email: lidd@nefu.edu.cn (D. Li), gcsui@nefu.edu.cn (G. Sui).

https://doi.org/10.1016/j.bbcan.2020.188410
Diterima 29 Juni 2020; Diterima dalam bentuk revisi 21 Juli 2020; Diterima 21 Juli 2020
Tersedia online 19 Agustus 2020
0304-419X / © 2020 Elsevier BV Semua hak dilindungi undang-undang.
W. Wang, dkk. BBA - Review tentang Cancer 1874 (2020) 188410
Gbr. 1. Struktur G-quadruplexes. A.Struktur kuartet G yang dibentuk oleh susunan koplanar empat guanin yang dipegang oleh pasangan basa Hoogsteen antara N1
dan O6 mereka (disorot dengan warna merah), dan distabilkan oleh kation logam univalen pusat (M +), dengan preferensi untuk monovalen kation dengan urutan K + >
Na+ > Li+. B dan C. Varietas topologi dari G-quadruplexes intramolekul dan antarmolekul. D. Daerah loop dari struktur G4. Kepala panah menunjukkan arah untaian
DNA; guanine (G) terlihat pada gambar dengan lingkaran biru.

tetrad), masing-masing terdiri dari empat guanin planar yang dihubungkan 2


oleh ikatan hidrogen Hoogsteen dan distabilkan oleh kation monovalen [12,33,40]. Dalam ulasan ini, kami merangkum penemuan, karakterisasi
struktural, fitur fungsional dan protein pengikat MYC G4, dan
(terutama K+) di pusatnya (Gbr. 1A) [21-24]. Baru-baru ini, sebuah studi G4
mendiskusikan ligan spesifik yang menargetkan struktur MYC G4 sebagai
ChIP-seq menggunakan BG4, sebuah antibodi yang mengenali struktur G4,
terapi kanker yang menguntungkan.
mengungkapkan sekitar 10.000 struktur G4 di promotor dan 5 'daerah yang
tidak diterjemahkan dari gen yang sangat ditranskripsikan dalam genom
manusia [25,26]. Selain itu, banyak penelitian menunjukkan bahwa struktur 2. Penemuan dan karakteristik struktural utama MYC G4
DNA khusus ini dapat langsung divisualisasikan dalam sel manusia oleh
antibodi spesifik G4 (BG4 dan 1H6) dan probe khusus. Secara fungsional, Struktur asam nukleat memainkan peran penting dalam mengatur
struktur G4 terlibat dalam berbagai proses biologis, termasuk pemeliharaan berbagai fungsi sel dan peristiwa biologis. Sekarang sudah mapan bahwa
telomer, transkripsi, replikasi DNA, terjemahan, stabilitas mRNA, dan DNA dapat membentuk berbagai struktur sekunder selain struktur heliks
banyak lagi [21,27-31]. Para promotor banyak gen mengandung struktur ganda tangan kanan dari DNA bentuk-B (B-DNA) [31]. Khususnya, urutan
G4. Di antara mereka, yang paling banyak dipelajari dan paling DNA kaya G spesifik dalam genom dapat dilipat menjadi struktur G4 yang
berkarakteristik adalah MYC G4 [32]. Yang penting, beberapa kelompok sangat stabil [21,22,41]. Dalam beberapa tahun terakhir, struktur G4 telah
baru-baru ini menemukan sejumlah besar ligan pengikat MYC G4, dipelajari secara lebih menyeluruh dalam kimia dan biologi, terutama
sebagian besar senyawa molekul kecil dan gelombang pasang, dengan karena struktur molekulnya yang khusus dan beragam (Gambar 1B dan C).
aplikasi potensial dalam terapi kanker [12,33-39]. Saat ini, penekanan Berdasarkan jumlah molekul DNA yang terlibat dalam pembentukan
transkripsi MYC melalui stabilisasi struktur G4 yang dimediasi ligan struktur, struktur G4 dapat dikelompokkan menjadi dua kategori utama:
merupakan rute yang menarik dalam pengobatan kanker intramolekul (disebut juga unimolekuler) G4 yang dibentuk oleh satu
molekul DNA, dan antarmolekul
W. Wang, dkk. BBA - Review tentang Cancer 1874 (2020) 188410
G4 dibentuk oleh dua atau lebih molekul DNA (Gambar 1B dan C) 3
[21,42,43]. Untuk kasus G4 intramolekul, motif yang dilestarikan adalah dichroism melingkar (CD), dan pendekatan resonansi magnetik nuklir,
G≥3N1-7G≥3N1- 7G≥3N1-7G≥3 (N mewakili nukleotida apapun). Jadi, promotor Yang et al. menunjukkan bahwa Pu27 dapat membentuk struktur G4
yang mengandung empat saluran G kontinyu, masing-masing terdiri dari beruntai paralel "propeller-type" yang dominan di tramolekul oleh G-tracts 2
setidaknya tiga guanin, dapat dengan mudah dilipat menjadi G4 sampai 5 pada ujung 3 'dari urutan [59]. Menariknya, studi pencetakan kaki
intramolekuler [21,41]. Namun, Shklover et al. mengusulkan bahwa struktur DMS dari Sun dan Hurley menunjukkan struktur G4 beruntai paralel yang
G4 antarmolekul mungkin hadir di daerah promotor gen [44]. Dailey dkk. melibatkan saluran-G 1 sampai 4 dalam sistem plasmid superkoil dalam
juga menunjukkan bahwa promotor yang mengandung lebih dari empat larutan air (Gbr. 2A dan C) [60,61]. Meskipun urutan Pu27 dari promotor
saluran G dapat membentuk beberapa kuartet diskrit yang tetap dalam MYC dapat membentuk tiga jenis struktur G4 secara in vitro, struktur G4
kesetimbangan dinamis satu sama lain [45]. Dalam studi in vitro baru-baru paralel tipe baling-baling intramolekul utama melibatkan saluran G 2-5 (G7-
ini menggunakan ukuran eksklusi chromato graphy dan uji NuPAGE, G9, G11-G13, G16-G18, dan G20-G22 ) dan berfungsi in vivo (Gbr. 2D)
Rauser dan Weinhold mengungkapkan bahwa struktur multi-timer G4 [19]. Memang, Ambrus et al. mensintesis oligonukleotida 22-nukleo
benar-benar diubah menjadi G4 monomer dengan perlakuan 150 mM pasang, dinamai Pu22, menggunakan urutan saluran-G 2-5 dengan G4,
NaOH diikuti dengan netralisasi menggunakanK + buffer[46]. Dengan G14 dan G23 diganti oleh T. Ketika diperiksa dengan resonansi magnetik
demikian, struktur G4 antarmolekul yang dibentuk oleh promotor dalam inti (NMR), Pu22 sebagian besar dapat membentuk konformasi G4 paralel
dua penelitian ini dapat dihasilkan dari kondisi mental percobaan yang intramolekul (Gbr. 2A dan E) [20]. Struktur ini terdiri dari tiga kuartet guanin
tidak optimal dari uji in vitro, meskipun kondisi eksperimental ini lazim dan tiga loop samping, dengan dua loop nukleotida tunggal dan satu loop
digunakan dalam penelitian G4. cleotida nu ganda, untuk menghubungkan empat untai guanin dan
Selanjutnya, menurut orientasi relatif untai DNA selama proses menjaga struktur tetap stabil (Gbr. 2E). Urutan 3′-flanking membentuk
pelipatan, struktur G4 dapat mengasumsikan konformasi paralel, konformasi fold back stacking yang stabil yang membatasi ujung atas
antiparalel, hibrid, dan tingkat tinggi (Gbr. 1B) [21,47,48]. Selain itu, struktur G4, sedangkan urutan 5′-mengapit menutup ujung bawah G4,
keragaman topologi struktur ini juga muncul dari jumlah G-kuartet, sifat dengan adenin yang ditumpuk sempurna ke dalam kuartet bawah [20].
loop, stoi chiometry dan polaritas untai, sudut torsi ikatan glikosidik, sifat Yang penting, struktur G4 ini stabil secara termodinamika, dengan suhu
empat alur, dan lokasi linker dan loop. (termasuk loop di agonal, lateral dan leleh yang serupa (> 85 ° C) dengan MYC Pu27 [20]. Konsisten dengan
baling-baling) (Gbr. 1D) [21,49,50]. Keragaman struktural ini membedakan struktur air yang dideteksi oleh NMR, Stump et al. baru-baru ini
struktur G4 satu sama lain dalam genom manusia, yang memungkinkan menyelesaikan struktur utama Pu22 MYC G4 dengan difraksi sinar-X pada
kita merancang ligan spesifik yang dapat mengenali dan mengikat struktur larutan 2,35 Å [62]. Antara struktur berair dan kristal, mereka menunjukkan
ini secara berbeda untuk memodulasi ekspresi gen yang sesuai. perbedaan besar dalam konformasi nukleotida mengapit 5 'dan 3', tetapi
Selanjutnya akan diringkas penemuan dan karakterisasi MYC G4 dalam tidak pada struktur pusat G-kuartet. Baru-baru ini, hasil penelitian ini
mengatur ekspresi gen MYC. didukung lebih lanjut oleh kelompok kami menggunakan CD, uji
pergeseran mobilitas elektroforetik (EMSA) dan penelitian pewarnaan DNA
perak [63-65]. Oleh karena itu, data ini sangat menyarankan bahwa NHE
2.1. Bukti in vitro struktur MYC G4
III1 dalam promotor MYC membentuk struktur G4 paralel intramolekul stabil
utama yang melibatkan saluran-G 2-5 secara in vitro.
Regulasi transkripsional dari gen MYC sangat kompleks dan
melibatkan empat promotor, termasuk P0, P1, P2 dan P3, dan dua lokasi
awal skrip trans (Gbr. 2A) [16,51-53] . Di antara promotor ini, sebagian 2.2. Bukti berbasis sel struktur MYC G4
besar transkrip diproduksi oleh promotor P1 dan P2, dengan 75-90%
transkrip MYC berasal dari promotor P2 [51-53]. Selain itu, dua elemen cis Pembentukan struktur MYC G4 secara in vitro dalam kondisi fisiologis
penting terletak di hulu promotor P1 dan P2, yang ditunjuk sebagai elemen telah dibuktikan dengan jelas. Selain itu, bukti dari berbagai kelompok
urutan hulu jauh (FUSE) dan NHE III 1, dan mengatur transkripsi MYC penelitian menunjukkan keberadaannya dalam sel hidup.
dengan cara yang memikat (Gbr. 2A) [16,51]. FUSE adalah elemen cruise Pertama, BG4 adalah antibodi yang direkayasa dengan spesifitas tinggi
control yang berfungsi sebagai sensor fisik dari transkripsi gen MYC yang dan afinitas pengikatan ke struktur G4, tetapi pengikatan tidak terdeteksi ke
sedang berlangsung, sedangkan elemen NHE III 1 terlibat dalam jepit rambut RNA, ssDNA atau dsDNA [27]. Hänsel-Hertsch dkk.
mengaktifkan dan membungkam transkripsi [16,51]. Banyak penelitian mengembangkan protokol G4 ChIP-seq menggunakan BG4 untuk
telah berfokus pada elemen NHE III 1 , karena mengatur 80-90% dari memetakan lokasi seluruh genom dari struktur G4 dalam kromatin sel
keseluruhan transkripsi MYC [16-18]. NHE III1 berisi urutan kaya purin 27- HaCaT, kera tinosit manusia diabadikan [25]. Data mereka memverifikasi
bp (Pu27) yang terletak -142 hingga -115 bp di hulu promotor P1. pengayaan struktur G4 dalam promotor MYC dalam sel HaCaT
Khususnya, Pu27 terdiri dari lima traktat G con sekutif; tiga dari saluran ini dibandingkan dengan keratinosit epi dermal manusia normal. Kedua,
terdiri dari empat guanin dan dua saluran terdiri dari tiga guanin (Gambar nukleolin (NCL) adalah fosfoprotein nukleol multi fungsi 100-kDa dengan
2A) [40,54], dalam menentukan potensi tingginya untuk membentuk ikatan selektif pada struktur G4 beruntai paralel, terutama MYC G4 [66,67].
struktur G4. Yang penting, NCL hanya mengikat pada struktur G4 dari promotor MYC
Sejak tahun 1998, Simonsson et al. pertama kali mengusulkan bahwa daripada bentuk ssDNA atau dsDNA [66,67]. Studi ini memberikan bukti
Pu27 yang kaya G dapat membentuk struktur G4 intramolekul dengan yang kuat tentang keberadaan struktur MYC G4 dalam konteks seluler
menggunakan antibodi G4 dan protein pengikat G4. Dalam beberapa
kuartet G antiparalel dalamK + larutan[55]. Struktur G4 terdiri dari tiga
tahun terakhir, probe fluorescent molekul kecil tertentu telah muncul
kuartet G yang terbentuk dari saluran-G 1, 2, 4 dan 5 yang dihubungkan
sebagai pendekatan alternatif untuk memvisualisasikan dan melacak
dengan loop diagonal pusat dan dua loop lateral. Namun, Siddiqui-Jain et
struktur G4 [21,30]. Misalnya, sensor "nyala" fluoresen yang peka terhadap
al. melaporkan bahwa wilayah ini dilipat menjadi setidaknya dua struktur
lingkungan mikro, disebut sebagai BTC f, dilaporkan oleh Panda et al.
G4 yang berbeda [19]. Dimetilsulfat (DMS) dapat memetilasi N7 guanin
dalam ss dan dsDNA, tetapi N7 dalam struktur G4 ini tidak dapat diakses karena kemampuannya untuk secara spesifik mendeteksi MYC Pu22 G4
oleh DMS karena pembentukan ikatan hidrogen Hoogsteen [56,57]. versus bentuk dsDNA Pu22 dengan peningkatan intensitas fluoresensi dan
Dengan demikian, uji footprinting DMS dapat mengidentifikasi guanosin pergeseran biru [68]. Yang penting, mikroskop pemindaian laser confocal
mana yang dapat diintegrasikan ke dalam struktur G4. Dengan (CLSM) dapat mendeteksi pewarnaan selektif oleh BTC f dalam inti sel
menggunakan uji ini, struktur MYC G4 mayor melibatkan saluran-G 2, 3, 4 HepG2 [68]. Kemudian, Chauhan et al. mensintesis dua fluorescent
dan 5, dan pembentukan G4 minor melibatkan saluran-G 1, 2, 4 dan 5 binaphthyl amines (ligan 8 dan 12) dengan dua cincin triazole. Ini memiliki
(Gbr. 2A, B dan D) [19]. Kedua struktur G4 ini juga ditunjukkan untuk afinitas yang lebih tinggi ke MYC G4 dan berinteraksi dengan kuartet G
membentuk struktur paralel tipe baling-baling intramolekul oleh kelompok ujung 3, mungkin melalui restrukturisasi pembatasan TAA-3 [69]. Selain itu,
lain (Gbr. 2A, B dan D) [58]. Demikian pula, menggunakan footprinting dua probe ini
DMS,
W. Wang, et al. BBA - Review tentang Cancer 1874 (2020) 188410
Gbr. 2. Struktur G4 promotor gen MYC. A. Struktur promotor dari gen MYC dan elemen terkait yang berbeda. Unsur-unsur ini termasuk urutan 27-mer (Pu27) dari
wilayah NHE III 1 di hulu promotor P1 dan urutan yang dimodifikasi: (1) MYC-1245 berisi G-traktat nomor 1, 2, 4, dan 5, dan Nomor saluran-G 3 diganti dengan “TTTT”;
(2) MYC-1234 berisi G-tracts nomor 1, 2, 3, dan 4; (3) MYC-2345 berisi G-tracts nomor 2, 3, 4, dan 5, dan mutasi G4 menjadi T; (4) Pu22 (4/14 / 23T) berisi G-tracts
nomor 2, 3, 4, dan 5, dan mutasi G-ke-T pada G4, G14 dan G23. MENJADI. Gambar skema struktur MYC-1245, MYC-1234, MYC-2345, dan Pu22 (4/14 / 23T) G4.
Kepala panah menunjukkan arah untai DNA 5'-ke-3 '; Guanin, adenin, dan timin (G, A, dan T) masing-masing ditunjukkan oleh lingkaran biru, kotak tertutup, dan kotak
terbuka.

dipamerkan fluoresensi "turn-on" tanggapan dengan MYC G4 dan diwarnai 4


inti dengan warna biru dalam sel [69]. Pada 2019, kelompok lain dua binaphthyl amine dan 4b, tetapi tidak 9CI, secara signifikan dapat
merancang probe guanosin fluoresen yang menembus sel, bernama ADG, menekan ekspresi MYC melalui menstabilkan struktur MYC G4 [68-72].
yang menunjukkan preferensi pengikatan ke MYC Pu22 G4 dibandingkan Secara signifikan, 9CI menunjukkan spesifisitas, afinitas, dan fluoresensi
dengan G4 dan dsDNA lainnya. ADG terikat ke dua terminal G-kuartet dari yang lebih tinggi daripada probe lainnya. Oleh karena itu, studi lebih lanjut
MYC Pu22 G4 dan berinteraksi dengan saluran kaya G yang berdekatan diperlukan untuk mengembangkan probe fluoresen molekuler kecil dengan
[70]. Menggunakan confocal micro scopy, ADG diamati menembus peningkatan spesifisitas, afinitas dan fluoresensi, serta kemampuan
membran sel dan terlokalisasi di inti sel dengan fluoresensi biru [70]. Selain menstabilkan struktur MYC G4, untuk aplikasi yang efektif dalam diagnosis
itu, Zhai et al. menghasilkan probe fluoresen menyala, 9CI, yang dan pengobatan kanker.
menampilkan lektivitas tinggi dan fluoresensi yang kuat untuk MYC Pu22 Secara keseluruhan, bukti yang cukup menunjukkan bahwa elemen
G4 in vitro dan dalam sel [71]. Secara mekanis, 9CI dapat mengikat erat ke NHE III1 dalam promotor MYC membentuk struktur G4 paralel intramolekul
loop T20-A22 dan pada kuartet G ujung 3. Khususnya, analisis spektral CD stabil utama baik in vitro maupun dalam sel hidup.
mengungkapkan bahwa 9CI tidak mengubah struktur topologi MYC G4
[71]. Yang penting, probe ini memiliki sitotoksisitas yang dapat diabaikan
dalam sel dan fluoresensi yang diaktivasi G4 hampir hilang dalam lisat sel 3. Peran fungsional MYC G4 dalam mengaturtranskripsi
[71]. Baru-baru ini, Deiana et al. mensintesis probe rotor light-up rakitan
khusus lokasi, 4b, dengan kekuatan pengikatan, sensitivitas, selektivitas, motifG4 di promotor atau sekitar situs inisiasi transkripsi (TSS)
dan stabilisasi yang menonjol untuk struktur MYC Pu22 G4 secara in vitro mengatur kemajuan transkripsi berbagai gen baik secara positif atau
[72]. Probe 4b ini juga terikat ke struktur MYC G4 melalui penumpukan negatif (Gbr. 3) [21,31,76,77]. Pertama, struktur G4 di hulu TSS gen dapat
eksternal. Dalam sel HeLa, 4b bisa terlokalisasi secara istimewa di memblokir transkripsi jika mereka mengganggu pengikatan RNA
kompartemen subnuklir yang kaya G4 [72]. Tiga kelompok tambahan juga polimerase II atau faktor transkripsi, tetapi mereka juga dapat mendorong
mengembangkan probe DAOTA-M2, IZFL-2 dan DDG1 untuk mendeteksi transkripsi dengan bertindak sebagai situs pengikatan untuk faktor
struktur MYC G4 secara in vitro [73-75]; bagaimanapun, aplikasinya untuk transkripsi tertentu atau protein pengikat G4, atau mempertahankan
memvisualisasikan struktur G4 dalam sel hidup tidak divalidasi. Secara keterbukaan. Konformasi DNA untai yang berlawanan dengan G4 (Gbr. 3A
keseluruhan, penelitian ini sangat menyarankan adanya struktur G4 di dan B) [31,63-65,76,78]. Kedua, struktur G4 di hilir TSS untai pengkodean
promotor MYC dalam sel hidup. Selain itu, BTC f, ADG, gen umumnya meningkatkan transkripsi karena perannya dalam
mendorong inisiasi ulang transkripsi. Sebaliknya bila ditempatkan
W. Wang, et al. BBA - Review on Cancer 1874 (2020) 188410
Gambar 3. Pembentukan struktur G4 dalam promotor dapat mempengaruhi transkripsi gen. Setelah inisiasi transkripsi, gelembung transkripsi yang terbentuk secara
instan dapat memungkinkan segmen untai tunggal membentuk G-quadruplexes. Struktur A dan B. G4 yang terbentuk di hulu TSS dalam untai pengkodean atau
templat dapat mengarah ke transkripsi yang diaktifkan atau ditekan. C dan D. Selama pemanjangan transkripsi, struktur G4 yang terbentuk di hilir TSS dalam untai
pengkodean atau templat meningkatkan inisiasi ulang transkripsi atau blok pengenalan RNA polimerase II. TSS: situs inisiasi transkripsi; TF: faktor transkripsi; panah
merah: transkripsi ditekan; panah hijau: transkripsi yang diaktifkan.

Gambar 4. Diagram skematis dari efek protein pengikat MYC G4. A, B dan C. Protein mempromosikan, menstabilkan dan melepaskan struktur MYC G4, masing-
masing.

hilir TSS dalam untai template, motif G4 umumnya bertindak sebagai 5


penekan transkripsi melalui faktor transkripsi yang menghalangi yang faktor protein dapat mengaktifkan transkripsi MYC melalui pengikatan
mengikat dsDNA atau memblokir pengenalan RNA polimerase II (Gbr. 3C langsung ke wilayah NHE III 1 , termasuk spesifisitas protein 1 (SP1),
dan D) [31,76,78,79] . Dengan demikian, efek keseluruhan struktur G4 protein pengikat asam nukleat jari seng tipe CCHC (CNBP) dan memiliki
pada ekspresi gen bergantung pada fungsi protein pengikat, seperti faktor ribonukleoprotein nuklir yang erogenous K ( hnRNP K) [51,54]. Sebagai
transkripsi, RNA polimerase II dan komponen lain dalam mesin transkripsi. faktor transkripsi yang penting, SP1 mengenali dan mengikat situs
Motif G4 pada untai cetakan dalam wilayah NHE III 1 terbentuk di hulu pengikatannya di NHE IIIuntai gandatranskripsi 1, dan mengaktifkan
TSS gen MYC, yang menunjukkan perannya dalam mengatur transkripsi skripgen MYC [51,80]. Baik CNBP dan hnRNP K juga mempromosikan
MYC. Studi sebelumnya menunjukkan bahwa beberapa ekspresi gen MYC melalui pengikatan pada untai kaya G dari NHE III 1 dan
untai kaya C komplementernya, masing-masing [51,81,82]. Berdasarkan
pengamatan tersebut, struktur G4 di wilayah NHE III 1 merepresi MYC
W. Wang, et al. BBA - ReviewCancer 1874 (2020) 188410
Transkripsi Genmelalui pemblokiran pengikatan faktor transkripsi tersebut. Tabel 1
Secara konsisten, Siddiqui-Jain et al. mendemonstrasikan bahwa mutasi Ringkasan protein pengikat MYC G4.
basis tunggal G-ke-A dalam wilayah Pu27 dapat menyebabkan mempromosikan re-inisiasi transkripsi, dengan efek keseluruhan dari
peningkatan aktivitas promotor MYC 3 kali lipat dalam garis sel tumor aktivasi gen MYC [87]. Chen et al. juga menunjukkan bahwa CNBP
melalui desta bilizing motif G4 [19]. Sebenarnya, mutasi individu dari manusia terikat pada NHE III 1 dan mengkatalisis transformasi topologi dari
semua 14 guanine yang terlibat dalam pembentukan struktur MYC G4 ssDNA menjadi struktur G4. Akibatnya, CNBP dapat menyebabkan
dapat secara signifikan merangsang aktivitas transkripsi promotor MYC penurunan sementara dan kemudian aktivasi transkripsi gen MYC dalam
[83]. Sejalan dengan pengamatan ini, pengobatan garis sel tumor dengan sel [90]. Yang penting, pengikatan CNBP ke NM23-H2, protein dengan
ligan kecil yang menstabilkan G4 dapat secara signifikan menurunkan aktivitas pelepasan G4, diperlukan untuk aktivasi transkripsional dari gen
transkripsi MYC [19,84]. Data ini sangat menyarankan bahwa struktur G4 MYC [90]. Secara mekanis, formasi G4 paralel yang dipromosikan CNBP
spesifik yang dibentuk di wilayah NHE III 1 dari promotor MYC berfungsi di wilayah NHE III1 dapat menjelaskan transkripsi yang direpresi
sebagai peredam transkripsi. sementara, sedangkan NM23-H2 yang direkrut CNBP menyelesaikan
struktur G4 dan menyebabkan aktivasi promotor MYC [90].
Protein pemacu G4 lainnya adalah nukleofosmin (NPM1), sebuah
4. Regulasi oleh faktor protein yang mengikat struktur MYC G4 Struktur
fosfoprotein bolak-balik cleocytoplasmic nu. NPM1 terutama terlokalisasi di
nukleoli, dan terlibat dalam pematangan dan ekspor ribosom, sentro
G4 saja tidak dapat mengatur proses seluler tetapi membutuhkan beberapa duplikasi, dan respons terhadap rangsangan stres [91-93].
protein yang terkait [21,85,86]. Dalam beberapa dekade terakhir, banyak Mutasi pada NPM1 C-terminal merupakan lesi genetik yang paling sering
protein telah diidentifikasi untuk menengahi transkripsi gen MYC melalui pada leukemia myeloid akut, dan menyebabkan translokasi dari nukleus ke
pengikatan, promosi, stabilisasi, atau pelepasan motif G4-nya. Pada sitoplasma karena sinyal ekspor nuklir yang dihasilkan oleh domain C-
bagian berikut, kita akan membahas mekanisme molekuler protein terminal yang tidak distabilkan [92,94-96]. Domain C-terminal dari NPM1
pengikat MYC G4 dalam mengatur transkripsi gen MYC. Protein ini dapat berikatan dengan ssDNA dan ssRNA secara istimewa di atas dsDNA,
dibagi menjadi tiga kelompok berdasarkan fungsinya, termasuk mendorong dengan urutannya masing-masing [97,98]. Terutama, 70 residu terakhirnya
pembentukan G4, menstabilkan, atau melepaskan struktur G4 (Gbr. 4 dan (NPM1-C70) secara istimewa mengikat DNA G4, dan mendorong
Tabel 1). pembentukan MYC G4 [99-101]. Data ini menunjukkan bahwa NPM1
menekan ekspresi gen MYC melalui pengikatan ke ssDNA NHE III 1 dan
4.1. Protein yang mendorong pembentukan MYC G4 mendorong pembentukan G4.

Di antara protein yang mendorong pembentukan struktur G4, nukleolin


4.2. Protein mengikat dan menstabilkan struktur MYC G4
(NCL) adalah salah satu yang paling banyak dipelajari. Seperti dijelaskan
di atas, NCL dapat secara selektif mengikat MYC G4 baik secara in vitro
maupun dalam sel. Gonza´lez dkk. pertama kali menunjukkan bahwa NCL Di antara protein dalam kelompok ini, adenosin deaminase yang
dapat mempromosikan pembentukan MYC G4 dan menstabilkan struktur bekerja pada RNA 1 (ADAR1) adalah enzim pengedit RNA untai ganda,
[66]. Mereka juga membuktikan RNA binding domain (RBDs) 3 dan 4, dan dan terlibat dalam berbagai proses biologis, termasuk deamina adenosin
domain arginine-glycine-glycine (RGG), bertanggung jawab untuk mengikat menjadi inosin, Interferensi RNA dan pematangan mikroRNA [102-104].
dan menstabilkan MYC G4 [67]. NCL panjang penuh yang diekspresikan Selain itu, ADAR1 juga merupakan protein pengikat Z-DNA klasik dengan
secara ektopik atau wilayah minimalnya menghambat transkripsi yang domain pengikatan Z-DNA yang sangat terkonservasi dan afinitas selektif
dimediasi oleh promotor MYC [66,67]. Secara mekanis, NCL dapat terhadap Z-DNA, yang merupakan DNA untai ganda kidal yang dibentuk
mempromosikan pembentukan MYC G4 di wilayah NHE III 1 dan menekan dalam urutan kaya G dan pirimidin / pirimidin / purin berulang, dan dapat
transkripsi MYC dengan menutupi situs pengikatan aktivator transkripsi, diinduksi olehnegatif [supercoiling105-108]. Kang et al. melaporkan bahwa
seperti SP1. domain pengikatan DNA Z ADAR1 manusia dapat langsung mengikat MYC
G4, dan menstabilkan konformasi untai paralelnya, yang menyebabkan
Tidak seperti peran NCL yang dikarakterisasi dengan baik, regulasi
MYC G4 oleh CNBP tampak membingungkan. CNBP pertama kali dikenali ekspresi gen MYC tertekan [109]. Studi ini memberikan wawasan baru
sebagai protein pengikat ssDNA yang kaya G dan pengatur potensial dari tentang peran biologis protein pengikat Z-DNA dalam mengatur proses
elemen pengatur sterol [87,88]. Pengikatan in vitro ke ssRNA yang kaya G yang dimediasi G4, seperti replikasi DNA, transkripsi, dan terjemahan.
Dengan demikian, menarik untuk mencari protein pengikat Z-DNA
juga dilaporkan [87,89]. Jadi, CNBP mengikat untai kaya-G dari NHE III 1
tambahan yang mengikat ke struktur G4, karena kesamaan dalam urutan
dan mengaktifkan transkripsi gen MYC. Namun, me chanisme molekuler
yang mendasari peraturan ini tidak diketahui. Borgognone dkk. melaporkan pembentuk struktur dan fungsi biologis dari struktur ini [108,109].
Protein penstabil G4 lainnya adalah mutan P53. P53 adalah penekan
bahwa ortolog CNBP di C. arenarum dapat mengikat ssDNA dari NHE III 1,
tumor yang paling sering bermutasi pada kanker manusia, dan
mendorong pelipatannya menjadi G4 paralel, dan kemudian terlepas dari
inaktivasinya memainkan peran penting dalam transformasi keganasan
struktur. Dalam hal ini, pengikatan CNBP dapat membuat struktur DNA
yang relatif terbuka untuk memungkinkan akses faktor transkripsi dan [110,111]. Sebagai faktor transkripsi, transkripsi gen yang diatur oleh P53
bergantung pada hubungannya dengan elemen pengikatnya pada
promotor target [110,112]. Dalam

Jenis protein pengikat G4 Protein domain pengikat G4 Referensi

Protein mempromosikan pembentukan G4 NCL RBD 3 dan 4 dan domain RGG di wilayah C-terminal [66,67] CNBP Kotak RGG di wilayah terminal N dasar [87,90]
NPM1 70-aa NPM1 C-terminal domain [99-101]
Protein mengikat dan menstabilkan struktur G4 LARK Domain RBM [121]
ADAR1 The Z-DNA mengikat domain [109] P53 mutan Wilayah C-terminal (aa 320-393) [119,120]
Protein melepaskan struktur G4 PIF1 Domain helikase yang dilestarikan (aa 206-620, PIF1-HD) [129–131] BLM Wilayah sayap-β dari domain RQC [135]
WRN Dua kunci aa (T1024 and T1086) within RQC domain [136]
DDX5 Not described [142]
NM23-H2 Not described [151,153,154]
PARP-1 The first zinc finger domain [155]

6
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

addition, P53 has high affinity for various noncanonical DNA struc tures, binding could impact gene transcription through associating with other
including G4 DNA, Holliday junctions, cruciforms, triplex DNA, T loops, transcription factors, such as nuclear factor Y (NF-Y), P63 and P73
stem-loops, DNA bulges and hemicatenated DNA [113,114]. In these [113,117,118]. Petr et al. reported that the C-terminal region (amino acids
processes, P53 interacts with multiple key regulators, including 320-393) of P53 was responsible for binding to the MYC G4 structure with
transcription factors [115,116]. P53 mutants mostly deficient in DNA high affinity [119]. P53 mutations were mostly identi fied in its DNA binding
domain, located in the middle region of the protein. Thus, most P53 7
mutants still retain their affinity for G4 structures. Quante et al. reported the 3′ ssDNA and the RQC domain to the G4 structure [78,137–139]. In
that a P53 mutant could bind and stabilize MYC G4 in vitro [120]. The data human cells, BLM, WRN and RECQ4 are of special interest due to their
indicated that mutated P53 in cancer cells may still regulate gene involvement in genetic disorders, including cancers and premature ageing
expression through modulating G4 structure stability. [140]. As the first discovered G4 helicase, BLM can unwind both
Niu et al. recently identified LARK, an RNA recognition motif (RRM, intermolecular and intramolecular G4 structures [78,141]. Lee dkk. reported
also known as RBM)-containing protein, as a protein to bind and sta bilize the interaction of BLM's RQC domain with MYC G4, and re vealed the
the MYC G4 structure in vitro [121]. However, whether LARK keeps these importance of the RQC β-wing region in binding and un winding the G4
properties in a cellular environment was not tested. Im portantly, the motif [135]. Another important member of this sub family is WRN, exhibiting
authors demonstrated that the RBM domain was indis pensable for the G4 similar functions to BLM. Ketkar et al. reported that the RQC domain of
binding activity of LARK, implicating the G4- binding potential of other WRN could impart a 2-fold binding preference to G4 compared to non-G4
proteins containing this conserved domain. DNA substrates [136]. Using the NMR approach, they also identified two
key amino acids, T1024 and T1086, within the RQC domain of WRN,
involved in G4 binding and unwinding activities [136].
4.3. Proteins unwinding the MYC G4 structure
Recently, Wu et al. reported an RNA helicase named Asp-Glu-Ala Asp
(DEAD)-box polypeptide 5 (DDX5, also called p68) that could be extremely
Most G4 structures exhibit inhibitory effects on DNA replication,
proficient at unwinding the MYC G4 structure [142]. As a prototypical
transcription and translation. Thus, proteins with G4-resolving activ ities can
member of the large ATP-dependent RNA helicases family, DDX5
promote these processes [21,22]. Helicases are molecular motors that
participates in a broad spectrum of biological processes, such as
open dsDNA during replication and transcription. Multiple helicases have
transcription, RNA processing (eg alternative splicing and RNA transport),
been reported to preferentially resolve noncanonical secondary structures,
DNA replication, and ribosome and miRNA biogenesis [143–145]. The
including G4 motifs, Holliday junctions and tri plexes [78,122–124].
DDX5 protein has a core helicase domain that is composed of two RecA-
Helicases are divided into six superfamilies, SF1 to SF6, based on protein
like domains known as domain 1 (D1) and D2. D1 can bind to ATP, and D2
sequence homology [78]. According to moving directions on DNA,
can recognize dsRNA structures [146,147]. DDX5 could also effectively
helicases can also be classified into type A (3′ to 5′) and type B (5′ to 3′)
unwind the MYC G4 and other in tramolecular G4 structures. However, its
[78]. All G4-helicases require a single-stranded tail at either the 3′ or 5′ end
unwinding activity did not need a ssDNA tail nor direct participation of ATP
to ensure their loading onto DNA substrates. Moreover, the G4 helicases
hydrolysis [142], indicating a distinct mechanism among known G4
act catalytically, and require hydrolysis of nucleotide triphosphates,
helicases so far. In MCF-7 cells, DDX5 was enriched at the MYC promoter
normally ATP, and Mg2+ ions [78,122]. and induced its transcription, which could be repressed by a G4-stabilizing
PIF1 is an ATP-dependent 5′ to 3′ DNA SF1 helicase and a member of compound, TMPyP4 [142].
a highly conserved protein family. It contributes to telomere elon gation, These data suggest that the helicases PIF1, BLM, WRN and DDX5 can
Okazaki fragment processing and genome stability maintenance [125– efficiently unwind MYC G4 structures in a catalytic fashion. In addition,
127]. Many studies demonstrated the activity of PIF1 in un winding dsDNA, multiple proteins can destabilize G4 structures in a non-cat alytic fashion,
double DNA/RNA hybrid strands and other secondary structures with unwinding processes independent of ATP hydro lysis. Unlike the
[85,127,128]. Sanders first reported that human PIF1 could bind and aforementioned helicases, these proteins resolving different types of G4
unwind the tetramolecular parallel G4 DNA structures in vitro, dependent DNA or RNA structures have not been extensively investigated so far.
on its conserved helicase domain [129]. Noticeably, PIF1 showed higher One of the proteins to disrupt G4 structures is NM23-H2, belonging to a
affinity for the G4 motif than that for ssDNA and dsDNA. In addition, prior to class of proteins involved in transcriptional activation, and ori ginally
unwinding G4 DNA structures, PIF1 needed to bind to an extended (> 10 identified as nucleotide diphosphate kinases [148,149]. In hu mans, NM23-
nucleotides) 5′ ssDNA tail but not G4 motifs [129]. Other groups reported H2 was first identified as a cancer metastasis inhibitor and has been
that PIF1 unfolds an in tramolecular parallel G4 structure, such as MYC G4 substantially studied in recent years due to its role in tran scriptionally
[130,131]. Im portantly, PIF1-catalyzed unfolding of G4 DNA is highly activating the MYC gene through binding the NHE III 1 element in vitro and
dependent on both substrate sequences and reaction conditions [131]. in a cellular environment [148–150]. However, the exact mechanism of
Another class of G4-helicases is the RecQ subfamily, comprising 3′ to NM23-H2 in promoting MYC gene transcription remains controversial. To
5′ DNA SF2 helicases conserved from prokaryotes to eukaryotes, with five date, ample evidence has demonstrated that NM23-H2 could preferentially
members, including RECQ1, RECQ4, RECQ5, Bloom syndrome protein bind the G- and C-rich single strands of NHE III 1, but not the dsDNA
(BLM), and Werner syndrome protein (WRN) [78,132,133]. Among these [151,152]. However, Thakur et al. re ported the activity of NM23-H2 in
helicases, BLM and WRN have all three conserved RecQ domains, binding to MYC G4 and then un winding to ssDNA [153], which was
including a helicase domain, a RecQ C-terminal (RQC) do main, and a confirmed by two other studies [151,154]. These data strongly suggested a
helicase-and-RNaseD C-terminal (HRDC) domain [78,134]. Among them, functional role of NM23-H2 in promoting MYC gene transcription through
the RQC domain is required for the G4-specific un winding activities of disrupting the G4 motif in NHE III 1. Thus, MYC G4 and NM23-H2
these helicases [78,135,136]. Strikingly, if the intramolecular G4 is followed interaction may be a potential intervening point to suppress MYC
by a single stranded region as short as 5- 10 nucleotides, BLM and WRN expression in cancer therapies.
helicases can unwind G4 in an ATP independent fashion, with cooperative PARP1, poly(ADP-ribose)polymerase-1, also regulates MYC gene
binding of the HRDC domain to transcription through its interaction with the G4 structure [155]. PARP1
protein is abundant in the chromatin of eukaryotic cells and has a high
affinity for damaged DNA. It becomes catalytically active upon binding to
DNA breaks, and then cleaves nicotinamide adenine dinu cleotide (NAD +)
into nicotinamide and ADP-ribose, the latter of which can be conjugated to
nuclear acceptor proteins, including X-ray repair cross complementing 1
(XRCC1), histones, and PARP1 itself for auto regulation [156–159]. The
enzyme activity of PARP1 is affected by its interaction with DNA, single-
and double-stranded breaks, non-B-DNA structures and others
[156,160,161]. PARP1 exhibits high in vitro
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

binding affinity to intramolecular G4 structures in the KRAS and c-KIT drugs, since it is discretely globular in nature and structurally different from
promoters, and also increases its catalytic activity when binding to G4 dsDNA. As the MYC G4 structure functions as a silencer element of
structures [162,163]. Additionally, Feketa et al. showed that PARP1 could transcription, ligands binding and stabilizing G4 can po tentially be used as
bind to the MYC G4 structure and convert it into a tran scriptionally active therapeutics to reduce MYC expression in cancer cells. Moreover, the
B-DNA form [155]. detailed NMR spectroscopic and crystallographic studies together with
computational analyses offered a clear under standing of the MYC G4
structure [20,62]. Using these approaches, many research groups
5. Ligands targeting the MYC G4 structure as clinical anticancer discovered a great variety of molecules with activities to bind and stabilize
therapeutics the MYC G4 structure, including quin dolines [164–167], cationic
porphyrins [19,84,168–171], imidazoles [35,172], quinoxalines [36],
G4 structures are attractive molecular targets in developing antic ancer carbazoles [173–175], berberine deriva tives [176,177], isaindigotone
derivatives [154], bisaryldiketene deri vatives [178], cyclic naphthalene
diimide derivatives [179], methylene blue derivatives [180], amido
phthalocyanines [181], and others [182–187].
With the help of computational analysis and chem-biological assays,
four basic models of MYC G4 binding to its ligands were proposed,
including external stacking, intercalating, groove/loop/backbone binding,
and central channel binding (Fig. 5) [188–190]. In the external stacking
model, ligands with an extended planar aromatic system containing
delocalized π-electrons can stack on the 5′ and/or 3′ term inal G-quartet(s)
mainly through π-π interactions, since the extended planar aromatic
system is similar to a G-quartet in size and shape, and it is a rigid flat or
twisted surface (Fig. 5A). In the intercalating model, ligands can insert into
the interspace of two G-quartets (Fig. 5B). In the groove/loop/backbone
model, the side chains of ligands interact with the grooves or loops through
hydrogen bonds, which also promote water solubility of the ligands. The
positively charged amino groups of the ligands increase their binding
affinity for the grooves and nega tively charged backbone phosphates
through electrostatic interactions (Fig. 5C). In the central channel binding 8
model, small ligands with long, positively charged side chains lying at the less popular than the other three models. Most reported ligands are prone
negative electrostatic center of a G-quartet can stabilize the G4 structure, to binding to MYC G4 in an external stacking-manner. In the following
similar to the role of monovalent cations, including K +, Na+ and NH4+ (Fig. sections, we discuss the anticancer activities and mechanisms of several
5D). For any ligand, it would be difficult for it to insert between two G- typical G4-stabilizing ligands, including small molecular drugs and
quartets of a G4 structure due to its high stability and rigidity, but relatively peptides, that have been classified according to their binding patterns with
easy to bind to the G-quartets at its two ends. Thus, the intercalator binding MYC G4 (Table 2 and Fig. 6). Furthermore, we assess the potential of
is these ligands as novel anticancer therapeutics.

5.1. Small molecular drugs

In the past decade, many small molecular drugs, both natural and
synthetic compounds, have been identified to bind and stabilize the MYC
G4 structure. To date, a number of natural compounds have been reported
to repress MYC expression through stabilizing its G4 structure, such as
Fonsecin B, alkaloids, piperine and flavonoid Quercetin [182,191–193].
However, their binding affinity and specificity to MYC G4 were much lower
than the reported synthetic compounds. Thus, we focus on discussing the
anticancer activities and underlying mechan isms of important synthetic
G4-stabilizing compounds in this section.
Interestingly, several synthetic drugs bind MYC G4 through either of
two additional models, specifically the groove/loop/backbone binding and
central channel binding models. However, as described above, external
stacking on terminal G-quartets is the primary model of MYC G4-ligand
complexes. Based on the stacking interactions of these li gands, we further
divide them into three groups, of which the ligands bind the 5′, 3′ and both
terminal G-quartets (Fig. 5A).

5.1.1. Compounds binding to both 5′ and 3′ terminal G-quartets of G4


structures
Various synthetic small molecules have been identified to bind and
stabilize the MYC G4 structure at the 5′ and 3′ terminal G-quartets of G4
structures (Fig. 6A). Brown et al. discovered GQC-05, a 9-dimethyla
minoethoxy substituted ellipticine compound, that could bind MYC G4 with
high affinity when analyzed by CD, competition dialysis and sur face
plasmon resonance (SPR) approaches. Importantly, through sta bilizing
MYC G4 to repress MYC gene transcription, GQC-05 could cause cell
death of Burkitt's lymphoma [194]. However, GQC-05 also showed low
affinity to G4 structures in the promoters of other genes, such as B cell
leukemia/lymphoma 2 (BCL2), platelet-derived growth factor A (PDGF-A)
and hypoxia-inducible factor 1-alpha (HIF-1α) [194]. It could also bind to a
G4 structure in the pre-mRNA of BCL-X to modulate its splicing process
[195]. The combinatorial use of GQC-05 with Navitoclax, a BCL2/BCL-XL
inhibitor, could cause remarkable cytotoxicity in acute myelocytic leukemia
(AML) cells, more pro nounced than that of each molecule alone, or
Navitoclax in combina tion with cytarabine or doxorubicin [196]. The data
suggest that the combination of G4-stabilizing agents with clinical
therapeutics is an

Fig. 5. Schematic diagrams of four basic binding


models of ligands with G4. A. The external stacking
model with ligands stacking to the 5′, 3′ or both
terminal G-quartets of a G4 structure. B. The inter
calating model with ligands inserted between G
quartets. C. The groove/loop binding model with
ligands binding to the groove and loop of a G4
structure. D. The central channel binding model with
the ligands staying in the center of a G4 structure.
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410
Table 2
Summary of MYC G4 ligands with antitumor activity
Types of ligands Target/binding models Compounds Tumor type/cell lines Pre-clinical/clinical stages References

Small molecular drugs Binding both 5′ and 3′ terminal G-quartets GQC-05 Burkitt's lymphoma (CA46) Pre-clinical [194] SYUIO-05 Burkitt's lymphoma (Ramos and CA46) Pre-clinical
[164]
7a4 Burkitt's lymphoma (Raji) Pre-clinical with in vivo efficacy [165]
19d, 22d Squamous cell Carcinoma (SiHa) Pre-clinical [154]
TZ1 Colorectal Adenocarcinoma (HCT116) Pre-clinical [197]
DC-34 Myeloma (L363) Pre-clinical [34]
IZTZ-1 Melanoma (B16) Pre-clinical with in vivo efficacy [35]
Binding to 5′ terminal G-quartet T-BFQs Lymphoma (Raji) Lung cancer (A549) Pre-clinical with in vivo efficacy [167]

Binding to 3′ terminal G-quartet negative breast cancer (4T1) Pre-clinical with in vivo effic acy [36] Cz-1 Cervical cancer (HeLa)
IZCZ-3 Squamous cell Carcinoma (SiHa) Pre-clinical with in vivo efficacy [172] QN-1 Triple- Pre-clinical [174] ANTP Cervical cancer (HeLa) Pre-clinical [37]
MYC G4/NCL CX-3543 Most of cancers Phase II [203,204] MYC G4/NM23-H2 Compound 37 Squamous cell Carcinoma (SiHa) Pre-clinical with in vivo efficacy
[205] Stauprimide Renal cancer (RXF 393) Pre-clinical with in vivo efficacy [206]
Peptides Binding both 3′ and 5′ terminal G-quartets TH3 Cervical cancer (HeLa) Pre-clinical [ 38] Binding groove/loop/backbone KR12C Breast cancer (MCF-7) Pre-clinical [39] Not
described α,ε-PLL Not described Not described [211]

optional and promising strategy in cancer therapies. 9


SYUIO-05, a quindoline derivative, was reported to preferentially cell apoptosis [197]. This study provides an innovative approach based on
stabilize MYC G4 with a negligible effect on telomeric G4. Thus, SYUIO 05 DNA nano-templates, which may serve as an efficient strategy to generate
could induce cell senescence and block cell proliferation through target-selective ligands in drug discovery.
repressing MYC gene transcription. Additionally, SYUIO-05 could dis rupt In 2018, Calabrese et al. identified a trifluoromethyl-substituted
the binding between NM23-H2 and MYC G4, leading to down regulated compound DC-34 from a library of benzofuran derivative analogs [34,198].
MYC expression [164]. Dai et al. used NMR to demonstrated a DC-34 exhibited strong affinity, remarkable binding affinity to MYC G4, and
stereometric structure with two SYUIO-05 molecules bound to top and could efficiently downregulate MYC gene expression in multiple myeloma
bottom G-quartets of MYC G4 [166]. cells [34]. Unlike other G4-stabilizing compounds, DC-34 did not bind
Later on, the same research group designed three highly specific MYC dsDNA and showed relatively weak affinity to the G4 structures of other
G4-stabilizing small molecules, 7a4, 19d and 22d. Among them, 7a4 is a tested oncogenes [34]. Analysis by NMR spec troscopy indicated that two
disubstituted quindoline derivative by introducing a second cationic amino DC-34 molecules could independently bind to the 5′ and 3′ terminal G-
side chain and 5-N-methyl group based on the structure of SYUIO-05 (Fig. quartets of MYC G4 [34]. Ample evidence from this group indicate that DC-
6A) [165]. Importantly, 7a4 exhibited better stabi lity, binding affinity, and 34 is a potential therapeutic in mye loma treatments.
selectivity to G4 structures (over dsDNA) than SYUIO-05 in vitro [165]. In Most recently, Wu et al. designed an imidazole-benzothiazole con
molecular docking studies, 7a4 retained its ability to bind to the two terminal jugate, IZTZ-1, based on the structures of QN-1 and 4l, two previously
G-quartets of MYC G4, as SYUIO-05 did; meanwhile, its crescent aromatic reported G4 ligands [35,36,199]. As predicted, the core imidazole
core also stacked onto two gua nine residues of both G-quartets. Through benzothiazole structure of IZTZ-1 could stack on both 5′- and 3′-term inal
this binding, the 5-N elec tropositive center of SYUIO-05 overlapped with G-quartets of MYC G4 [35]. Meanwhile, the two extended posi tively
the cation channel of G4, acting as a monovalent cation, such as K + to charged 1-methylpiper-azine groups could increase the binding affinity of
stabilize MYC G4 [165]. Moreover, 7a4 has two amino acid side chains IZTZ-1 with negatively charged phosphate backbone via electrostatic
forming a V-shape and stretching toward two grooves. Thus, the complex interactions. Through these interactions, IZTZ-1 could strongly stabilize the
model greatly increases the binding stability, affinity and selectivity of 7a4 MYC G4 structure [35]. Consistently, IZTZ-1 could reduce the proliferation
to MYC G4. Functionally, 7a4 could decrease MYC gene expression and migration of melanoma cells, and suppress tumor growth in a
through stabilizing its G4 structure and disrupting NM23-H2/G4 interaction melanoma mouse model through downregulating MYC expression [35]. In
in Burkitt's lymphoma cells. This anticancer activity was also verified in a addition, IZTZ-1 showed a desirable solubility and its core structure
tumor xenograft mouse model [165]. In addition to quindoline deri vatives, a possessed potential drug-likeness, suggesting its great potential as a
series of new isaindigotone derivatives were also designed. Among them, molecular drug in clinical treatment of melanoma.
19d and 22d could specifically bind to MYC G4 and prevent its interaction
with NM23-H2-G4 [154]. As for the binding model, these two molecules
5.1.2. Compounds binding to 5′ terminal G-quartets of G4 structures
could bind to MYC G4 with similar end stacking interactions [154].
Moreover, the two phenyl groups of their structures were twisted, which Several highly specific MYC G4 stabilizing molecules have been reported
further induced two cationic amine side chains to bind the negative to exclusively bind the 5′ terminal G-quartets of G4 structures (Fig. 6B). In
phosphate backbone through electrostatic interactions [154]. Likewise, 19d 2017, Zeng et al. designed a series of 11-triazole benzo furoquinoline
and 22d also exhibited significant anticancer activity in SiHa cells [154]. derivatives (T-BFQs) based on the chemical structure of quindoline [167].
Panda et al. used magnetic nanoparticle-linked MYC G4 as a tem plate Three of these compounds, 5a, 5h and 5l, exhibited the highest binding
for covalently assembling potent azide-alkyne partners to gen erate high- affinity to MYC G4, but could still bind to other G4 structures [167]. Notably,
affinity G4 ligands. They identified a triazole compound Tz1 (Fig. 6A) with these compounds could significantly pro mote the formation of parallel MYC
higher affinity for and better stabilization of MYC G4 over dsDNA than other G4 structures in the absence of cations, indicating their strong interaction
triazole derivatives [197]. Importantly, in col orectal adenocarcinoma with MYC G4. In line with the results of the biochemical studies, the three
HCT116 cells, Tz1 could specifically inhibit MYC gene transcription through compounds could specifically inhibit MYC in human lymphoma cells
stabilizing its G4 structure, leading to through stabilizing MYC G4 [167]. In addition, 5l could significantly reduce
tumor growth in a lung cancer xenograft mouse model [167]. Nevertheless,
the spe cificity of these molecules to MYC G4 versus other G4 structures
has not
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410
Fig. 6. Chemical structures of compounds that directly or indirectly target MYC G4 structure. A. Compounds binding to both 5′ and 3′ terminal G-quartets of MYC G4
structure, including GQC-05, SYUIO-05, 7a4, 19d, 22d, Tz1, DC-34 and IZTZ-1. B. Compounds binding to 5′ terminal G-quartet of MYC G4 structure, including T BFQs,
IZCZ-3 and QN-1. C. Chemical structures of compounds binding to 3′ terminal G-quartet of MYC G4 structure, including Cz-1 and ANTP. D. Compounds indirectly
targeting MYC G4 structure, including CX-3543, stauprimide and compound 37.
10
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

been explored. 11
In 2018, Hu et al. synthesized a four-leaf clover-like ligand, IZCZ-3, ability of ANTP to G4 structures increased its potential selectivity of G4
which was designed through structural modification of aryl-substituted DNA over dsDNA. Moreover, ANTP-mediated MYC G4 stabilization was
imidazole/carbazole conjugates. IZCZ-3 could preferentially bind and clearly verified by fluorescence titration, CD titration and melting, and Taq
stabilize MYC G4 structure in vitro [172]. The molecular docking study polymerase stop assays [37]. Furthermore, ANTP exhibited marked
indicated that IZCZ-3 could perfectly stack on the 5′ terminal G-quartet plane of cytotoxicity in HeLa and HEK293T cells at 5.3 μM, but not in primary
G4 via π−π interaction. Moreover, the positively charged central imidazole moiety NIH3T3 and HDFa cells at a concentration up to 100 μM, indicating its
of IZCZ-3 was located in the ion channel of MYC G4, contributing to its strong G4 excellent cancer cell selectivity [37].
binding affinity [172]. Likewise, a line of in vitro cell-based studies also Taken together, the above data from different groups suggest that
proved that IZCZ-3 could ef fectively downregulate MYC transcription synthetic small molecules can inhibit MYC transcription by directly binding
through specifically targeting its G4 structure, leading to growth inhibition of the G4 in the MYC promoter using these three binding models, and
SiHa cells, but not normal BJ fibroblasts and primary mouse mesangial consequently stabilizing its structure, leading to significant antic ancer
cells [172]. Fur thermore, IZCZ-3 exhibited a strong inhibitory activity of effects in cancer cells and/or in mouse models. However, several
repressing tumor growth in nude mice xenografted with human cervical compounds can repress MYC transcription and inhibit cancer cell growth
squamous cancer cells [172]. Thus, IZCZ-3 is a promising anticancer ligand through indirectly stabilizing the MYC G4 structure. As men tioned above,
due to its high selectivity to MYC G4 structure. NCL and NM23-H2 mediate MYC G4 folding or unfolding. Indeed, small
Compared with IZCZ-3, a quinoline derivative, QN-1, showed rela tively molecules specifically targeting the NM23-H2-G4 inter action, or inducing
high selectivity to MYC G4 with several advantages, such as a drug-like NCL redistribution from nucleoli into the nucleo plasm were reported. For
core structure, a new promising scaffold targeting MYC G4, and ease in example, a fluoroquinolone derivative, CX 3543, also known as quarfloxin,
chemical synthesis [36]. After examination by different approaches, is the first-in-class G4-interacting compound to reach the human phase II
including absorption titrations, CD analysis, NMR titrations and 2- clinical trial for neuroendo crine/carcinoid tumors [203]. CX-3543 could bind
aminopurine (2-Ap) fluorescent titrations experiments, QN-1 was identified to ribosomal DNA (rDNA) G4 and selectively disrupt its interaction with
as the most promising G4-stabilizing ligand with high se lectivity to MYC NCL, thereby inducing NCL relocation into nucleoplasm, which could inhibit
G4 through stacking onto the 5′-end G-quartet, and also binding to the DNA Polymerase I-mediated transcription and induce cancer cell apoptosis
grooves or loops with its two outstretched N-methyl piperazine side chains [203]. After being relocated in nucleoplasm, NCL may also promote MYC
[36]. Consistently, QN-1 showed a decent an ticancer activity against triple- G4 formation and thus inhibit MYC transcription [203,204]. In addition,
negative breast cancer (TNBC) in both cell-based and mouse model several compounds repress MYC transcription through dis rupting the
studies. Importantly, QN-1-caused MYC repression was significantly interaction between NM23-H2 and MYC G4, such as a novel isaindigotone
greater than that of other G4-mediated genes, such as BCL2, c-KIT, VEGF derivative named compound 37 and stauprimide (Fig. 6D) [205,206]. The
and HRAS [36]. Moreover, the TNBC 4T1 cells with overexpressed MYC anticancer activity of compound 37 was at tributed to its high specific
were more sensitive to QN-1, as compared to normal cells. Of note, by binding affinity to NM23-H2 protein, re sulting in effectively disrupting the
comparing to classical TNBC chemo therapeutics (eg doxorubicin, interaction of NM23-H2 with G4 and greatly decreasing MYC transcription
paclitaxel and cisplatin) and a pan G4-binding molecule BRACO19, 2.5 μM [205]. However, stauprimide could indirectly disrupt NM23-H2 and G4
of QN-1 could completely repress the growth of 4T1 cells, but the other interaction through blocking NM23-H2 nuclear translocation [206].
compounds at this con centration could only eliminate less than 70% of the Therefore, disruption of MYC G4 interaction with its binding protein, such
cells [36]. These data indicate that QN-1 is an alternative and promising as NCL and NM23-H2, re presents an alternative strategy to develop novel
agent in the treatment of TNBC. anticancer ther apeutics.

5.1.3. Compounds binding to 3′ terminal G-quartets of G4 structures 5.2. Peptides


Several compounds bind only to the 3′ terminal G-quartet to stabi lize the
G4 structure (Fig. 6C). Das et al. synthesized a series of car bazole Although a large number of small molecules targeting MYC G4 were
derivatives that could bind and stabilize MYC G4 [174]. Among these reported, most of them have not been used in clinical applications due to
compounds, Cz-1 exhibited prominent G4 versus dsDNA se lectivity, their off-target effects and narrow therapeutic windows [207]. In view of
especially to MYC G4. Cell-based experiments indicated that Cz-1 could these disadvantages, peptides can offer an alternative and promising
downregulate MYC expression through its binding and sta bilizing MYC G4, approach in designing G4-targeted anticancer therapeutics based on their
and consequently promote HeLa cell apoptosis [174]. As a fluorescent variability and plasticity of 3D structures, and potential in recapitulating
probe, Cz-1 is mainly localized in the nucleus with blue fluorescence [174]. G4/protein interactions. These features can help re searchers to design
When detected by the G4 antibody BG4 in an im munocytochemical study, peptides with increased intracellular affinity and selectivity in targeting G4
Cz-1 treated cells showed increased immuno positive foci compared to the structures, and relatively low off-target ef fects in regulating oncogene
control cells, colocalized with the Cz-1 signal [27]. Cz-1 treatment showed expression [39]. In addition, peptides also have advantages in clinical
more BG4 foci than those in the control cells, and Cz-1 colocalized with applications, including lower im munogenicity, minimal drug-drug
BG4-stained sites [174]. The results confirmed that Cz-1 could stabilize interaction, easy intra-tumoral dif fusion, fast blood clearance, and
and/or promote the forma tion of G4s in cancer cells. Therefore, this new excellent tolerability in patients [39,208]. So far, several peptides have
modulable carbazole scaffold exhibits high G4 binding affinity and been reported to bind and sta bilize the MYC G4 structure.
stabilizing activity with potent anticancer potential. In 2018, Dutta et al. synthesized a crescent-shaped thiazole tripep tide,
Another example is ANTP, an anthraquinone with a tri-N-methyl pyrrole named TH3, with selective binding affinity for MYC G4 over other
side chain, which was synthesized by Roy et al. through con jugating investigated G4s or dsDNA [38]. Furthermore, the NMR analysis in dicated
anthraquinone pharmacophore with oligopyrrole carbox amides, and that the extended planar structure of TH3 could interact with MYC G4
showed remarkable binding affinity to various G4 DNA structures, through binding to both 5′ and 3′ terminal G-quartets [38]. The AT-rich
especially MYC G4 [37]. The discovery of this compound was based on capping structures of two terminal G-quartets are unique in MYC G4, and
efficient π-π interaction of anthraquinone derivatives with G-quartets and thus determine TH3's specificity for the MYC G4 structure over other G4
the capability of oligopyrrole moieties in binding to the grooves of dsDNA structures. Consistently, TH3 significantly inhibited MYC gene transcription
and G4 DNA [37,200–202]. This dual recognition through a G4-dependent mechanism, and caused S phase arrest and
apoptosis of HeLa cells [38], suggesting its potential as
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

an anticancer agent. apoptotic signaling through the E2F1/VEGF-A/ BCL2 axis in MCF-7 cells
Also in 2018, Sengupta et al. designed a peptide, KR12C, by pruning [39]. Overall, the KR12C peptide could spe cifically target MYC G4 and
the G4-binding domain (FK13) of a naturally occurring human Cathelicidin exhibit significant anticancer activity.
(LL37) protein [39,209,210]. KR12C could interact with MYC G4 in a In addition to these two G4-stabilizing peptides, Marzano et al. re cently
groove/loop/backbone binding model, and selectively stabilize the 5′- reported a synthetic analogue of ε-poly-l-lysine (ε-PLL), named as α,ε-PLL,
propeller loop of MYC G4 via arginine-driven electro static-interactions at binding to both MYC G4 and telomeric G4 structures with high affinity [211].
the sugar-phosphate backbone [39]. The treatment of KR12C could reduce The ε-PLL peptide is a cationic biopolymer isolated from the marine
the binding of SP1 and NM23-H2 to the NHE III 1 region, suggesting its bacterium Bacillus subtilis with antibacterial and anticancer activity, and is
activity in stabilizing MYC G4. Consistently, KR12C could promote used worldwide as a food preservative [211,212]. Im portantly, both ε-PLL
and α,ε-PLL are used in the biomedical applica tions, such as helpers of 12
drug and gene delivery [213,214]. Thus, de veloping more specific peptides vast library of natural compounds. Finally, we need to evaluate the
based on amino acid sequences of α,ε PLL peptide and evaluating its anticancer activities and side-effects of G4-stabilizing ligands in com
anticancer activity in cancer cells are interesting research topics in future binatorial use with clinical chemotherapeutics to synergistically elim inate
studies. cancer cells. Collectively, the role of G4 structures in MYC reg ulation
Notably, multiple G4-unwinding proteins have been reported to bind provides fertile questions for future research.
and unfold the MYC G4 structure, leading to MYC gene activation and
malignant transformation. Hence, it is a logical strategy in dis covering Author contributions
anticancer peptides through mapping the shortest binding domain of a G4-
binding protein and synthesizing peptides based on its sequence to WW, DL and GS conceived and wrote the manuscript. DBS critically
competitively disrupt the protein-G4 complex. read the manuscript and provided constructive suggestions. WW designed
the tables and created the figures. SH, YG and YY searched and organized
6. Conclusions and future perspectives literature. Semua penulis telah membaca dan menyetujui versi naskah
yang diterbitkan.
Cancer initiation and progression are often accompanied by altered
gene expression. Significantly, the MYC gene has acquired a formidable Declaration of Competing Interest
oncogenic reputation based on its aberrant expression through elevated
and constitutive transcription, leading to unrestricted proliferation, impaired The authors declare no conflict of interest.
apoptosis, enhanced angiogenesis and metastasis, and genomic instability
in a variety of cancers. Among the regulatory ele ments in the MYC gene,
Acknowledgments
the GC-rich NHE III1 region upstream of its P1 promoter has been identified
as the most critical regulator of its ex pression. Ample biochemical, cell-
This work was supported by the Fundamental Research Funds for the
based and in vivo evidence discussed in this review suggested that the GC-
Central Universities (2572017BA10) to DL, the National Natural Science
rich NHE III1 element in the MYC promoter is able to form an intramolecular Foundation of China (81802798) to DL and (81872293 and 81672795) to
parallel G4 structure. The MYC G4 functions as a transcription repressor GS, the China Postdoctoral Science Foundation (2018M631897) and the
motif in MYC gene ex pression. The biological role of MYC G4 depends on Heilongjiang Postdoctoral Fund (LBH-Z16005) to DL.
its interacting proteins that may promote G4 formation, stabilize their
structures, or unwind them. Stabilization of G4 structures by specific
References
ligands can affect their interaction with proteins leading to reduced MYC
tran scription. These ligands are thus potential anticancer molecules.
Especially, based on well characterized MYC G4, small molecules and [1] T. Ngoma, World Health Organization cancer priorities in developing countries, Ann.
Oncol. 17 (Suppl. 8) (2006) viii9–viii14.
peptides can be designed to target this structure and potentially used as [2] G. De Rosa, D. De Stefano, A. Galeone, Oligonucleotide delivery in cancer therapy,
promising anticancer drugs through downregulating MYC gene ex pression. Expert Opin. Obat Deliv. 7 (2010) 1263–1278.
Although important progress has been made in targeting MYC G4, we [3] D. Hanahan, RA Weinberg, Hallmarks of cancer: the next generation, Cell 144 (2011)
646–674.
propose several prospective routes for future basic and clinical in
[4] EY Lee, WJ Muller, Oncogenes and tumor suppressor genes, Cold Spring Harb.
vestigations. First, the development of additional probes with high Perspect. Biol. 2 (2010) a003236.
specificity, affinity and fluorescence to detect MYC G4 elements in living [5] CY Lin, J. Loven, PB Rahl, RM Paranal, CB Burge, JE Bradner, TI Lee, RA Young,
cells will expand the experimental tools available for studying these Transcriptional amplification in tumor cells with elevated c-Myc, Cell 151 (2012) 56–67.
[6] CV Dang, MYC on the path to cancer, Cell 149 (2012) 22–35. [7] A. Kiessling, B. Sperl,
important structures. Second, future studies should seek to iden tify A. Hollis, D. Eick, T. Berg, Selective inhibition of c-Myc/Max dimerization and DNA binding
additional proteins that bind and stabilize MYC G4; we predict that these by small molecules, Chem. Biol. 13 (2006) 745–751.
insights will reveal novel therapeutic targets. Third, future work should also [8] ZE Stine, ZE Walton, BJ Altman, AL Hsieh, CV Dang, MYC, metabolism, and cancer,
Cancer Discov. 5 (2015) 1024–1039.
design novel, stabilizing ligands with high specificity and affinity for MYC G4 [9] KA O'Donnell, EA Wentzel, KI Zeller, CV Dang, JT Mendell, c-Myc-regulated microRNAs
and/or its associated proteins. We predict these molecules will possess modulate E2F1 expression, Nature 435 (2005) 839–843. [10] J. van Riggelen, A. Yetil, DW
significant anticancer activity and low side-ef fects in the clinic. Fourth, it is Felsher, MYC as a regulator of ribosome biogenesis and protein synthesis, Nat. Rev. Cancer
10 (2010) 301–309.
important to screen naturally occurring small molecules as G4-binding and [11] TR Kress, A. Sabo, B. Amati, MYC: connecting selective transcriptional control to global
oncogene silencing agents from a RNA production, Nat. Rev. Cancer 15 (2015) 593–607.
[12] BL Allen-Petersen, RC Sears, Mission possible: advances in MYC therapeutic
targeting in cancer, Biodrugs 33 (2019) 539–553.
[13] NM Sodir, LB Swigart, AN Karnezis, D. Hanahan, GI Evan, L. Soucek, Endogenous
Myc maintains the tumor microenvironment, Genes Dev. 25 (2011) 907–916.
[14] D. Annibali, JR Whitfield, E. Favuzzi, T. Jauset, E. Serrano, I. Cuartas, S. Redondo-
Campos, G. Folch, A. Gonzalez-Junca, NM Sodir, D. Masso-Valles, ME Beaulieu, LB
Swigart, MM Mc Gee, MP Somma, S. Nasi, J. Seoane, GI Evan, L. Soucek, Myc
inhibition is effective against glioma and reveals a role for Myc in proficient mitosis,
Nat. Komun. 5 (2014) 4632.
[15] Y. Li, SC Casey, DW Felsher, Inactivation of MYC reverses tumorigenesis, J. Intern.
Med. 276 (2014) 52–60.
[16] I. Wierstra, J. Alves, The c-myc promoter: still MysterY and challenge, Adv. Res kanker.
99 (2008) 113–333.
[17] TL Davis, AB Firulli, AJ Kinniburgh, Ribonucleoprotein and protein factors bind to an H-
DNA-forming c-myc DNA element: possible regulators of the c-myc gene, Proc. Natl.
Acad. Sci. USA 86 (1989) 9682–9686.
[18] EH Postel, SE Mango, SJ Flint, A nuclease-hypersensitive element of the human c-myc
promoter interacts with a transcription initiation factor, Mol. Sel. Biol. 9 (1989) 5123–
5133.
[19] A. Siddiqui-Jain, CL Grand, DJ Bearss, LH Hurley, Direct evidence for a G quadruplex in
a promoter region and its targeting with a small molecule to repress c-MYC transcription,
Proc. Natl. Acad. Sci. USA 99 (2002) 11593–11598. [20] A. Ambrus, D. Chen, J. Dai, RA
Jones, D. Yang, Solution structure of the
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

biologically relevant G-quadruplex element in the human c-MYC promoter. Implications structures mark human regulatory chromatin, Nat. Genet. 48 (2016) 1267–1272.
for G-quadruplex stabilization, Biochemistry 44 (2005) 2048–2058. [21] D. Varshney, J. [26] R. Hansel-Hertsch, J. Spiegel, G. Marsico, D. Tannahill, S. Balasubramanian, Genome-
Spiegel, K. Zyner, D. Tannahill, S. Balasubramanian, The regula tion and functions of DNA wide mapping of endogenous G-quadruplex DNA structures by chromatin
and RNA G-quadruplexes, Nat. Rev. Mol. Biol Sel. 21 (2020) 459–474. immunoprecipitation and high-throughput sequencing, Nat. Protoc. 13 (2018) 551–564.
[22] CK Kwok, CJ Merrick, G-quadruplexes: prediction, characterization, and bio logical [27] G. Biffi, D. Tannahill, J. McCafferty, S. Balasubramanian, Quantitative visualiza tion of
application, Trends Biotechnol. 35 (2017) 997–1013. DNA G-quadruplex structures in human cells, Nat. Chem. 5 (2013) 182–186.
[23] Z. Tan, YH Hao, KW Zheng, Kinetics, conformation, stability, and targeting of G [28] A. Henderson, Y. Wu, YC Huang, EA Chavez, J. Platt, FB Johnson, RM Brosh Jr., D.
quadruplexes from a physiological perspective, Biochem. Biofis. Res. Komun. (2020), Sen, PM Lansdorp, Detection of G-quadruplex DNA in mammalian cells, Nucleic Acids
https://doi.org/10.1016/j.bbrc.2020.04.047. Res. 42 (2014) 860–869.
[24] A. Virgilio, A. Russo, T. Amato, G. Russo, L. Mayol, V. Esposito, A. Galeone, [29] HY Liu, Q. Zhao, TP Zhang, Y. Wu, YX Xiong, SK Wang, YL Ge, JH He, P. Lv, TM Ou,
Monomolecular G-quadruplex structures with inversion of polarity sites: new topologies JH Tan, D. Li, LQ Gu, J. Ren, Y. Zhao, ZS Huang, Conformation selective antibody enables
and potentiality, Nucleic Acids Res. 45 (2017) 8156–8166. genome profiling and leads to discovery of parallel G quadruplex in human telomeres, Cell
[25] R. Hansel-Hertsch, D. Beraldi, SV Lensing, G. Marsico, K. Zyner, A. Parry, M. Di Antonio, Chem. Biol. 23 (2016) 1261–1270.
J. Pike, H. Kimura, M. Narita, D. Tannahill, S. Balasubramanian, G quadruplex [30] P. Chilka, N. Desai, B. Datta, Small molecule fluorescent probes for G- quadruplex
visualization as potential cancer theranostic agents, Molecules 24 (2019). [31] ML Bochman, [
K. Paeschke, VA Zakian, DNA secondary structures: stability and function of G-quadruplex
structures, Nat. Rev. Genet. 13 (2012) 770–780. [32] R. Rigo, M. Palumbo, C. Sissi, G-
quadruplexes in human promoters: a challenge for therapeutic applications, Biochim. Biofis. 4
Acta Gen. Subj. 1861 (2017) 1399–1413.
[33] BJ Chen, YL Wu, Y. Tanaka, W. Zhang, Small molecules targeting c-Myc on cogene:
promising anti-cancer therapeutics, Int. J. Biol. Sci. 10 (2014) 1084–1096. [34] DR Calabrese, 8
X. Chen, EC Leon, SM Gaikwad, Z. Phyo, WM Hewitt, S. Alden, TA Hilimire, F. He, AM
Michalowski, JK Simmons, LB Saunders, S. Zhang, D. Connors, KJ Walters, BA Mock, JS
Schneekloth Jr., Chemical and structural studies provide a mechanistic basis for recognition ]
of the MYC G-quadruplex, Nat. Komun. 9 (2018) 4229.
[35] TY Wu, Q. Huang, ZS Huang, MH Hu, JH Tan, A drug-like imidazole-ben zothiazole
conjugate inhibits malignant melanoma by stabilizing the c-MYC G quadruplex, S
Bioorg. Chem. 99 (2020) 103866.
[36] MH Hu, TY Wu, Q. Huang, G. Jin, New substituted quinoxalines inhibit triple negative
breast cancer by specifically downregulating the c-MYC transcription, Nucleic Acids .
Res. 47 (2019) 10529–10542.
[37] S. Roy, A. Ali, M. Kamra, K. Muniyappa, S. Bhattacharya, Specific stabilization of
promoter G-Quadruplex DNA by 2,6-disubstituted amidoanthracene-9,10-dione based A
dimeric distamycin analogues and their selective cancer cell cytotoxicity, Eur. J. Med.
Chem. 195 (2020) 112202.
[38] D. Dutta, M. Debnath, D. Muller, R. Paul, T. Das, I. Bessi, H. Schwalbe, J. Dash, Cell s
penetrating thiazole peptides inhibit c-MYC expression via site-specific tar geting of c-
MYC G-quadruplex, Nucleic Acids Res. 46 (2018) 5355–5365.
a
[39] P. Sengupta, N. Banerjee, T. Roychowdhury, A. Dutta, S. Chattopadhyay, S. Chatterjee,
Site-specific amino acid substitution in dodecameric peptides de termines the stability
and unfolding of c-MYC quadruplex promoting apoptosis in cancer cells, Nucleic Acids
m
Res. 46 (2018) 9932–9950.
[40] LH Hurley, DD Von Hoff, A. Siddiqui-Jain, D. Yang, Drug targeting of the c-MYC
promoter to repress gene expression via a G-quadruplex silencer element, Semin. it
Oncol. 33 (2006) 498–512.
[41] R. Hansel-Hertsch, M. Di Antonio, S. Balasubramanian, DNA G-quadruplexes in the
human genome: detection, functions and therapeutic potential, Nat. Rev. Mol. Biol Sel. s
18 (2017) 279–284.
[42] DL Ma, Z. Zhang, M. Wang, L. Lu, HJ Zhong, CH Leung, Recent developments in G-
quadruplex probes, Chem. Biol. 22 (2015) 812–828. u
[43] WF Yuan, LY Wan, H. Peng, YM Zhong, WL Cai, YQ Zhang, WB Ai, JF Wu, The
influencing factors and functions of DNA G-quadruplexes, Cell Biochem. Funct. 38
(2020) 524–532. ,
[44] J. Shklover, P. Weisman-Shomer, A. Yafe, M. Fry, Quadruplex structures of muscle gene
promoter sequences enhance in vivo MyoD-dependent gene expression, Nucleic Acids
Res. 38 (2010) 2369–2377. M
[45] MM Dailey, MC Miller, PJ Bates, AN Lane, JO Trent, Resolution and char acterization of
the structural polymorphism of a single quadruplex-forming se quence, Nucleic Acids
Res. 38 (2010) 4877–4888. .
[46] V. Rauser, E. Weinhold, Quantitative formation of monomeric G-quadruplex DNA from
multimeric structures of c-Myc promoter sequence, ChemBioChem (2020),
https://doi.org/10.1002/cbic.202000159. T
[47] DJ Patel, AT Phan, V. Kuryavyi, Human telomere, oncogenic promoter and 5'- UTR G-
quadruplexes: diverse higher order DNA and RNA targets for cancer therapeutics,
Nucleic Acids Res. 35 (2007) 7429–7455. a

i,

it

a
, 3

Y Perspectives for applying G-quadruplex structures in neurobiology and neuro


pharmacology, Int. J. Mol. Sci. 20 (2019).
[49] T. Vy Thi Le, S. Han, J. Chae, HJ Park, G-quadruplex binding ligands: from naturally
. occurring to rationally designed molecules, Curr. Pharm. Des. 18 (2012) 1948–1972.
[50] HJ Lipps, D. Rhodes, G-quadruplex structures: in vivo evidence and function, Trends
Cell Biol. 19 (2009) 414–422.
I [51] TA Brooks, LH Hurley, The role of supercoiling in transcriptional control of MYC and its
importance in molecular therapeutics, Nat. Rev. Cancer 9 (2009) 849–861.
[52] LA Carabet, PS Rennie, A. Cherkasov, Therapeutic inhibition of Myc in cancer. Structural
m bases and computer-aided drug discovery approaches, Int. J. Mol. Sci. 20 (2018).
[53] N. Meyer, LZ Penn, Reflecting on 25 years with MYC, Nat. Rev. Cancer 8 (2008) 976–
990.
a [54] V. Gonzalez, LH Hurley, The c-MYC NHE III(1): function and regulation, Annu. Rev.
Pharmacol. Toksikol. 50 (2010) 111–129.
[55] T. Simonsson, P. Pecinka, M. Kubista, DNA tetraplex formation in the control re gion of c-
i, myc, Nucleic Acids Res. 26 (1998) 1167–1172.
[56] W. Huang, PJ Smaldino, Q. Zhang, LD Miller, P. Cao, K. Stadelman, M. Wan, B. Giri, M.
Lei, Y. Nagamine, JP Vaughn, SA Akman, G. Sui, Yin Yang 1 contains G-quadruplex
H
structures in its promoter and 5'-UTR and its expression is modu lated by G4 resolvase
1, Nucleic Acids Res. 40 (2012) 1033–1049.
[57] B. Onel, G. Wu, D. Sun, C. Lin, D. Yang, Electrophoretic mobility shift assay and
.
dimethyl sulfate footprinting for characterization of G-quadruplexes and G quadruplex-protein
complexes, Methods Mol. Biol. 2035 (2019) 201–222.
K [58] AT Phan, YS Modi, DJ Patel, Propeller-type parallel-stranded G-quadruplexes in the
human c-myc promoter, J. Am. Chem. Soc. 126 (2004) 8710–8716. [59] D. Yang, LH Hurley,
Structure of the biologically relevant G-quadruplex in the c MYC promoter, Nucleosides
a Nucleotides Nucleic Acids 25 (2006) 951–968. [60] D. Sun, LH Hurley, The importance of
negative superhelicity in inducing the formation of G-quadruplex and i-motif structures in the
c-Myc promoter: im plications for drug targeting and control of gene expression, J. Med.
s Chem. 52 (2009) 2863–2874.
[61] RI Mathad, E. Hatzakis, J. Dai, D. Yang, c-MYC promoter G-quadruplex formed at the 5'-
end of NHE III1 element: insights into biological relevance and parallel stranded G-quadruplex
h stability, Nucleic Acids Res. 39 (2011) 9023–9033.
[62] S. Stump, TC Mou, SR Sprang, NR Natale, HD Beall, Crystal structure of the major
quadruplex formed in the promoter region of the human c-MYC oncogene, PLoS One
i 13 (2018) e0205584.
[63] T. Yan, B. Zhao, Q. Wu, W. Wang, J. Shi, D. Li, DB Stovall, G. Sui, Characterization of
G-quadruplex formation in the ARID1A promoter, Int. J. Biol. Macromol. 147 (2020)
w 750–761.
[64] X. Zhang, B. Zhao, T. Yan, A. Hao, Y. Gao, D. Li, G. Sui, G-quadruplex structures at the
promoter of HOXC10 regulate its expression, Biochim. Biofis. Acta Gene Regul. Mech.
a 1861 (2018) 1018–1028.
[65] Y. Li, X. Zhang, Y. Gao, J. Shi, L. Tang, G. Sui, G-quadruplexes in the BAP1 pro moter
positively regulate its expression, Exp. Cell Res. 369 (2018) 147–157. [66] V. Gonzalez, K.
g Guo, L. Hurley, D. Sun, Identification and characterization of nucleolin as a c-myc G-
quadruplex-binding protein, J. Biol. Chem. 284 (2009) 23622–23635.
[67] V. Gonzalez, LH Hurley, The C-terminus of nucleolin promotes the formation of the c-
i, MYC G-quadruplex and inhibits c-MYC promoter activity, Biochemistry 49 (2010) 9706–
9714.
[68] D. Panda, M. Debnath, S. Mandal, I. Bessi, H. Schwalbe, J. Dash, A nucleus-ima ging
N probe that selectively stabilizes a minor conformation of c-MYC G-quadruplex and
down-regulates c-MYC transcription in human cancer cells, Sci. Rep. 5 (2015) 13183.
[69] A. Chauhan, R. Paul, M. Debnath, I. Bessi, S. Mandal, H. Schwalbe, J. Dash, Synthesis
. of fluorescent binaphthyl amines that bind c-MYC G-quadruplex DNA and repress c-MYC
expression, J. Med. Chem. 59 (2016) 7275–7281.
[70] P. Saha, YP Kumar, T. Das, D. Muller, I. Bessi, H. Schwalbe, J. Dash, G-quad ruplex-
S specific cell-permeable guanosine-anthracene conjugate inhibits telomere elongation
and induces apoptosis by repressing the c-MYC gene, Bioconjug. Chem. 30 (2019)
3038–3045.
h [71] Q. Zhai, C. Gao, J. Ding, Y. Zhang, B. Islam, W. Lan, H. Hou, H. Deng, J. Li, Z. Hu, HI
Mohamed, S. Xu, C. Cao, SM Haider, D. Wei, Selective recognition of c-MYC Pu22 G-
quadruplex by a fluorescent probe, Nucleic Acids Res. 47 (2019) 2190–2204.
i
[72] M. Deiana, K. Chand, J. Jamroskovic, RN Das, I. Obi, E. Chorell, N. Sabouri, A site-
specific self-assembled light-up rotor probe for selective recognition and sta bilization of c-
MYC G-quadruplex DNA, Nanoscale 12 (2020) 12950–12957.
o
[73] A. Kotar, B. Wang, A. Shivalingam, J. Gonzalez-Garcia, R. Vilar, J. Plavec, NMR
structure of a triangulenium-based long-lived fluorescence probe bound to a G
quadruplex, Angew. Chem. Int. Ed. Eng. 55 (2016) 12508–12511.
d
[74] MH Hu, J. Zhou, WH Luo, SB Chen, ZS Huang, R. Wu, JH Tan, Development of a smart
fluorescent sensor that specifically recognizes the c-MYC G-quadruplex, Anal. Chem.
a 91 (2019) 2480–2487.
[75] Y. Pavan Kumar, P. Saha, D. Saha, I. Bessi, H. Schwalbe, S. Chowdhury, J. Dash,
Fluorescent dansyl-guanosine conjugates that bind c-MYC promoter G-quadruplex and
, downregulate c-MYC expression, ChemBioChem 17 (2016) 388–393.
[76] P. Armas, A. David, NB Calcaterra, Transcriptional control by G-quadruplexes: in vivo
roles and perspectives for specific intervention, Transcription 8 (2017) 21–25.
1 [77] Y. Qin, LH Hurley, Structures, folding patterns, and functions of intramolecular DNA G-
quadruplexes found in eukaryotic promoter regions, Biochimie 90 (2008)
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

1149–1171. 101 (2004) 6140–6145.


[78] O. Mendoza, A. Bourdoncle, JB Boule, RM Brosh Jr., JL Mergny, G-quad ruplexes [84] AT Phan, V. Kuryavyi, HY Gaw, DJ Patel, Small-molecule interaction with a five-
and helicases, Nucleic Acids Res. 44 (2016) 1989–2006. guanine-tract G-quadruplex structure from the human MYC promoter, Nat. Chem.
[79] N. Kim, The interplay between G-quadruplex and transcription, Curr. Med. Chem. 26 Biol. 1 (2005) 167–173.
(2019) 2898–2917. [85] ZY Sun, XN Wang, SQ Cheng, XX Su, TM Ou, Developing novel G-quad ruplex ligands:
[80] B. Majello, P. De Luca, G. Suske, L. Lania, Differential transcriptional regulation of c-myc from interaction with nucleic acids to interfering with nucleic acid (-)protein interaction,
promoter through the same DNA binding sites targeted by Sp1-like proteins, Oncogene Molecules 24 (2019).
10 (1995) 1841–1848. [86] V. Brazda, L. Haronikova, JC Liao, M. Fojta, DNA and RNA quadruplex-binding proteins,
[81] M. Takimoto, T. Tomonaga, M. Matunis, M. Avigan, H. Krutzsch, G. Dreyfuss, D. Int. J. Mol. Sci. 15 (2014) 17493–17517.
Levens, Specific binding of heterogeneous ribonucleoprotein particle protein K to the human [87] M. Borgognone, P. Armas, NB Calcaterra, Cellular nucleic-acid-binding protein, a
c-myc promoter, in vitro, J. Biol. Chem. 268 (1993) 18249–18258. transcriptional enhancer of c-Myc, promotes the formation of parallel G-quad ruplexes,
[82] EF Michelotti, T. Tomonaga, H. Krutzsch, D. Levens, Cellular nucleic acid binding Biochem. J. 428 (2010) 491–498.
protein regulates the CT element of the human c-myc protooncogene, J. Biol. Chem. [88] TB Rajavashisth, AK Taylor, A. Andalibi, KL Svenson, AJ Lusis, Identification of a zinc
270 (1995) 9494–9499. finger protein that binds to the sterol regulatory element, Science 245 (1989) 640–643.
[83] CL Grand, TJ Powell, RB Nagle, DJ Bearss, D. Tye, M. Gleason-Guzman, LH Hurley, [89] D. Benhalevy, SK Gupta, CH Danan, S. Ghosal, HW Sun, HG Kazemier, K. Paeschke,
Mutations in the G-quadruplex silencer element and their relationship to c-MYC M. Hafner, SA Juranek, The human CCHC-type zinc finger nucleic acid-binding protein
overexpression, NM23 repression, and therapeutic rescue, Proc. Natl. Acad. Sci. USA binds G-rich elements in target mRNA coding sequences and promotes translation,
Cell Rep. 18 (2017) 2979–2990. 14
[90] S. Chen, L. Su, J. Qiu, N. Xiao, J. Lin, JH Tan, TM Ou, LQ Gu, ZS Huang, D. Li, [105] PR Marshall, Q. Zhao, X. Li, W. Wei, A. Periyakaruppiah, EL Zajaczkowski, LJ Leighton,
Mechanistic studies for the role of cellular nucleic-acid-binding protein (CNBP) in SU Madugalle, D. Basic, Z. Wang, J. Yin, WS Liau, A. Gupte, CR Walkley, TW Bredy,
regulation of c-myc transcription, Biochim. Biofis. Acta 1830 (2013) 4769–4777. Dynamic regulation of Z-DNA in the mouse prefrontal cortex by the RNA-editing
[91] S. Grisendi, C. Mecucci, B. Falini, PP Pandolfi, Nucleophosmin and cancer, Nat. Rev. enzyme Adar1 is required for fear extinction, Nat. Neurosci. 23 (2020) 718–729.
Cancer 6 (2006) 493–505. [106] S. Bae, D. Kim, KK Kim, YG Kim, S. Hohng, Intrinsic Z-DNA is stabilized by the
[92] CG Grummitt, FM Townsley, CM Johnson, AJ Warren, M. Bycroft, Structural conformational selection mechanism of Z-DNA-binding proteins, J. Am. Chem. Soc.
consequences of nucleophosmin mutations in acute myeloid leukemia, J. Biol. Chem. 133 (2011) 668–671.
283 (2008) 23326–23332. [107] H. Jiao, L. Wachsmuth, S. Kumari, R. Schwarzer, J. Lin, RO Eren, A. Fisher, R. Lane,
[93] B. Falini, C. Mecucci, E. Tiacci, M. Alcalay, R. Rosati, L. Pasqualucci, R. La Starza, D. GR Young, G. Kassiotis, WJ Kaiser, M. Pasparakis, Z-nucleic-acid sen sing triggers
Diverio, E. Colombo, A. Santucci, B. Bigerna, R. Pacini, A. Pucciarini, A. Liso, M. Vignetti, P. ZBP1-dependent necroptosis and inflammation, Nature 580 (2020) 391–395.
Fazi, N. Meani, V. Pettirossi, G. Saglio, F. Mandelli, F. Lo-Coco, PG Pelicci, MF Martelli, [108] Y. Zhang, Y. Cui, R. An, X. Liang, Q. Li, H. Wang, H. Wang, Y. Fan, P. Dong, J. Li, K.
Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype, N. Cheng, W. Wang, S. Wang, G. Wang, C. Xue, M. Komiyama, Topologically
Engl. J. Med. 352 (2005) 254–266. constrained formation of stable Z-DNA from normal sequence under physiological
[94] MT Werner, C. Zhao, Q. Zhang, MA Wasik, Nucleophosmin-anaplastic lym phoma conditions, J. Am. Chem. Soc. 141 (2019) 7758–7764.
kinase: the ultimate oncogene and therapeutic target, Blood 129 (2017) 823–831. [109] HJ Kang, TV Le, K. Kim, J. Hur, KK Kim, HJ Park, Novel interaction of the Z DNA
[95] B. Falini, MP Martelli, N. Bolli, P. Sportoletti, A. Liso, E. Tiacci, T. Haferlach, Acute binding domain of human ADAR1 with the oncogenic c-Myc promoter G quadruplex, J.
myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity? Blood Mol. Biol. 426 (2014) 2594–2604.
117 (2011) 1109–1120. [110] A. Hafner, ML Bulyk, A. Jambhekar, G. Lahav, The multiple mechanisms that regulate
[96] B. Falini, P. Sportoletti, MP Martelli, Acute myeloid leukemia with mutated NPM1: p53 activity and cell fate, Nat. Rev. Mol. Biol Sel. 20 (2019) 199–210. [111] VJN Bykov, SE
diagnosis, prognosis and therapeutic perspectives, Curr. Opin. Oncol. 21 (2009) 573– Eriksson, J. Bianchi, KG Wiman, Targeting mutant p53 for efficient cancer therapy, Nat. Rev.
581. Cancer 18 (2018) 89–102.
[97] D. Wang, A. Baumann, A. Szebeni, MO Olson, The nucleic acid binding activity of [112] IM Fingerman, SD Briggs, p53-mediated transcriptional activation: from test tube to
nucleolar protein B23.1 resides in its carboxyl-terminal end, J. Biol. Chem. 269 (1994) cell, Cell 117 (2004) 690–691.
30994–30998. [113] V. Brazda, M. Fojta, The rich world of p53 DNA binding targets: the role of DNA
[98] TS Dumbar, GA Gentry, MO Olson, Interaction of nucleolar phosphoprotein B23 with structure, Int. J. Mol. Sci. 20 (2019).
nucleic acids, Biochemistry 28 (1989) 9495–9501. [114] V. Brazda, J. Coufal, Recognition of local DNA structures by p53 protein, Int. J. Mol.
[99] L. Federici, A. Arcovito, GL Scaglione, F. Scaloni, C. Lo Sterzo, A. Di Matteo, B. Falini, Sci. 18 (2017).
B. Giardina, M. Brunori, Nucleophosmin C-terminal leukemia-associated domain [115] G. Sui, B. Affarel, Y. Shi, C. Brignone, NR Wall, P. Yin, M. Donohoe, MP Luke, D. Calvo,
interacts with G-rich quadruplex forming DNA, J. Biol. Chem. 285 (2010) 37138– SR Grossman, Y. Shi, Yin Yang 1 is a negative regulator of p53, Cell 117 (2004) 859–
37149. 872.
[100] PL Scognamiglio, C. Di Natale, M. Leone, M. Poletto, L. Vitagliano, G. Tell, D. Marasco, [116] JP Kruse, W. Gu, Modes of p53 regulation, Cell 137 (2009) 609–622. [117] S.
G-quadruplex DNA recognition by nucleophosmin: new insights from protein dissection, Weissmueller, E. Manchado, M. Saborowski, JPT Morris, E. Wagenblast, CA Davis, SH
Biochim. Biofis. Acta 1840 (2014) 2050–2059. Moon, NT Pfister, DF Tschaharganeh, T. Kitzing, D. Aust, EK Markert, J. Wu, SM Grimmond,
[101] A. Gallo, C. Lo Sterzo, M. Mori, A. Di Matteo, I. Bertini, L. Banci, M. Brunori, L. Federici, C. Pilarsky, C. Prives, AV Biankin, SW Lowe, Mutant p53 drives pancreatic cancer metastasis
Structure of nucleophosmin DNA-binding domain and analysis of its complex with a G- through cell-autono mous PDGF receptor beta signaling, Cell 157 (2014) 382–394.
quadruplex sequence from the c-MYC promoter, J. Biol. Chem. 287 (2012) 26539– [118] MH Stindt, PA Muller, RL Ludwig, S. Kehrloesser, V. Dotsch, KH Vousden, Functional
26548. interplay between MDM2, p63/p73 and mutant p53, Oncogene 34 (2015) 4300–4310.
[102] SH Roth, EY Levanon, E. Eisenberg, Genome-wide quantification of ADAR adenosine- [119] M. Petr, R. Helma, A. Polaskova, A. Krejci, Z. Dvorakova, I. Kejnovska, L. Navratilova,
to-inosine RNA editing activity, Nat. Methods 16 (2019) 1131–1138. [103] Q. Jiang, J. Isquith, M. Adamik, M. Vorlickova, M. Brazdova, Wild-type p53 binds to MYC promoter G-
MA Zipeto, RH Diep, J. Pham, N. Delos Santos, E. Reynoso, J. Chau, H. Leu, E. Lazzari, E. quadruplex, Biosci. Rep. 36 (2016).
Melese, W. Ma, R. Fang, M. Minden, S. Morris, B. Ren, G. Pineda, F. Holm, C. Jamieson, [120] T. Quante, B. Otto, M. Brazdova, I. Kejnovska, W. Deppert, GV Tolstonog, Mutant p53 is
Hyper-editing of cell-cycle regulatory and tumor suppressor RNA promotes malignant a transcriptional co-factor that binds to G-rich regulatory regions of active genes and
progenitor propagation, Cancer Cell 35 (2019) 81–94 (e87). generates transcriptional plasticity, Cell Cycle 11 (2012) 3290–3303.
[104] H. Chung, JJA Calis, X. Wu, T. Sun, Y. Yu, SL Sarbanes, VL Dao Thi, AR Shilvock, HH [121] K. Niu, L. Xiang, Y. Jin, Y. Peng, F. Wu, W. Tang, X. Zhang, H. Deng, H. Xiang, S. Li, J.
Hoffmann, BR Rosenberg, CM Rice, Human ADAR1 pre vents endogenous RNA Wang, Q. Song, Q. Feng, Identification of LARK as a novel and conserved G-
from triggering translational shutdown, Cell 172 (2018) 811–824 (e814). quadruplex binding protein in invertebrates and vertebrates, Nucleic Acids Res. 47
(2019) 7306–7320.
[122] M. Sauer, K. Paeschke, G-quadruplex unwinding helicases and their function in vivo,
Biochem. Soc. Trans. 45 (2017) 1173–1182.
[123] MC Whitby, L. Ryder, RG Lloyd, Reverse branch migration of Holliday junctions by
RecG protein: a new mechanism for resolution of intermediates in re combination and
DNA repair, Cell 75 (1993) 341–350.
[124] M. Guo, K. Hundseth, H. Ding, V. Vidhyasagar, A. Inoue, CH Nguyen, R. Zain, JS Lee,
Y. Wu, A distinct triplex DNA unwinding activity of ChlR1 helicase, J. Biol. Chem. 290
(2015) 5174–5189.
[125] VP Schulz, VA Zakian, The saccharomyces PIF1 DNA helicase inhibits telomere
elongation and de novo telomere formation, Cell 76 (1994) 145–155. [126] ME Budd, CC Reis,
S. Smith, K. Myung, JL Campbell, Evidence suggesting that Pif1 helicase functions in DNA
replication with the Dna2 helicase/nuclease and DNA polymerase delta, Mol. Sel. Biol. 26
(2006) 2490–2500.
[127] K. Paeschke, JA Capra, VA Zakian, DNA replication through G-quadruplex motifs is
promoted by the Saccharomyces cerevisiae Pif1 DNA helicase, Cell 145 (2011) 678–
691.
[128] T. George, Q. Wen, R. Griffiths, A. Ganesh, M. Meuth, CM Sanders, Human Pif1
helicase unwinds synthetic DNA structures resembling stalled DNA replication forks,
Nucleic Acids Res. 37 (2009) 6491–6502.
[129] CM Sanders, Human Pif1 helicase is a G-quadruplex DNA-binding protein with G
quadruplex DNA-unwinding activity, Biochem. J. 430 (2010) 119–128. [130] AK Byrd, KD
Raney, A parallel quadruplex DNA is bound tightly but unfolded slowly by pif1 helicase, J. Biol.
Chem. 290 (2015) 6482–6494.
[131] AK Byrd, MR Bell, KD Raney, Pif1 helicase unfolding of G-quadruplex DNA is highly
dependent on sequence and reaction conditions, J. Biol. Chem. 293 (2018) 17792–
17802.
[132] JM Caruthers, DB McKay, Helicase structure and mechanism, Curr. Opin. Struct. Biol.
12 (2002) 123–133.
[133] RJ Monnat Jr., Human RECQ helicases: roles in DNA metabolism, mutagenesis and
cancer biology, Semin. Cancer Biol. 20 (2010) 329–339.
[134] DL Croteau, V. Popuri, PL Opresko, VA Bohr, Human RecQ helicases in DNA repair,
recombination, and replication, Annu. Pdt. Biochem. 83 (2014) 519–552. [135] S. Lee, AR
Lee, KS Ryu, JH Lee, CJ Park, NMR investigation of the interaction between the RecQ C-
terminal domain of human bloom syndrome protein and G quadruplex DNA from the human c-
Myc promoter, J. Mol. Biol. 431 (2019)
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

794–806. [141] H. Sun, JK Karow, ID Hickson, N. Maizels, The Bloom's syndrome helicase un winds G4
[136] A. Ketkar, M. Voehler, T. Mukiza, RL Eoff, Residues in the RecQ C-terminal do main of DNA, J. Biol. Chem. 273 (1998) 27587–27592.
the human werner syndrome helicase are involved in unwinding G quadruplex DNA, J. [142] G. Wu, Z. Xing, EJ Tran, D. Yang, DDX5 helicase resolves G-quadruplex and is involved
Biol. Chem. 292 (2017) 3154–3163. in MYC gene transcriptional activation, Proc. Natl. Acad. Sci. USA 116 (2019) 20453–
[137] JB Budhathoki, S. Ray, V. Urban, P. Janscak, JG Yodh, H. Balci, RecQ-core of BLM 20461.
unfolds telomeric G-quadruplex in the absence of ATP, Nucleic Acids Res. 42 (2014) [143] RM Nyamao, J. Wu, L. Yu, X. Xiao, FM Zhang, Roles of DDX5 in the tumor igenesis,
11528–11545. proliferation, differentiation, metastasis and pathway regulation of human malignancies,
[138] S. Chatterjee, J. Zagelbaum, P. Savitsky, A. Sturzenegger, D. Huttner, P. Janscak, ID Biochim. Biofis. Acta Rev. Cancer 1871 (2019) 85–98.
Hickson, O. Gileadi, E. Rothenberg, Mechanistic insight into the interaction of BLM [144] W. Cai, Z. Xiong Chen, G. Rane, S. Satendra Singh, Z. Choo, C. Wang, Y. Yuan, T. Zea
helicase with intra-strand G-quadruplex structures, Nat. Komun. 5 (2014) 5556. Tan, F. Arfuso, CT Yap, LS Pongor, H. Yang, MB Lee, B. Cher Goh, G. Sethi, T. Benoukraf,
[139] P. Janscak, PL Garcia, F. Hamburger, Y. Makuta, K. Shiraishi, Y. Imai, H. Ikeda, TA V. Tergaonkar, A. Prem Kumar, Wanted DEAD/H or alive: helicases winding up in cancers, J.
Bickle, Characterization and mutational analysis of the RecQ core of the bloom Natl. Cancer Inst. 109 (2017).
syndrome protein, J. Mol. Biol. 330 (2003) 29–42. [145] A. Mazurek, W. Luo, A. Krasnitz, J. Hicks, RS Powers, B. Stillman, DDX5 regulates DNA
[140] JK Karow, L. Wu, ID Hickson, RecQ family helicases: roles in cancer and aging, Curr. replication and is required for cell proliferation in a subset of breast cancer cells,
Opin. Genet. Dev. 10 (2000) 32–38. Cancer Discov. 2 (2012) 812–825.
[146] AL Mallam, M. Del Campo, B. Gilman, DJ Sidote, AM Lambowitz, Structural basis for 15
RNA-duplex recognition and unwinding by the DEAD-box helicase Mss116p, Nature quadruplexes in a plasmid using atomic force microscopy, Biochemistry 51 (2012)
490 (2012) 121–125. 578–585.
[147] V. Hashemi, A. Masjedi, B. Hazhir-Karzar, A. Tanomand, SS Shotorbani, M. Hojjat- [162] VA Soldatenkov, AA Vetcher, T. Duka, S. Ladame, First evidence of a functional
Farsangi, G. Ghalamfarsa, G. Azizi, E. Anvari, B. Baradaran, F. Jadidi Niaragh, The interaction between DNA quadruplexes and poly(ADP-ribose) polymerase-1, ACS
role of DEAD-box RNA helicase p68 (DDX5) in the development and treatment of Chem. Biol. 3 (2008) 214–219.
breast cancer, J. Cell. Physiol. 234 (2019) 5478–5487. [163] S. Cogoi, M. Paramasivam, A. Membrino, KK Yokoyama, LE Xodo, The KRAS promoter
[148] EH Postel, SJ Berberich, SJ Flint, CA Ferrone, Human c-myc transcription factor PuF responds to Myc-associated zinc finger and poly(ADP-ribose) polymerase 1 proteins,
identified as nm23-H2 nucleoside diphosphate kinase, a candidate suppressor of which recognize a critical quadruplex-forming GA-element, J. Biol. Chem. 285 (2010)
tumor metastasis, Science 261 (1993) 478–480. 22003–22016.
[149] EH Postel, SJ Berberich, JW Rooney, DM Kaetzel, Human NM23/nucleoside [164] TM Ou, J. Lin, YJ Lu, JQ Hou, JH Tan, SH Chen, Z. Li, YP Li, D. Li, LQ Gu, ZS Huang,
diphosphate kinase regulates gene expression through DNA binding to nuclease Inhibition of cell proliferation by quindoline derivative (SYUIQ-05) through its
hypersensitive transcriptional elements, J. Bioenerg. Biomembr. 32 (2000) 277–284. preferential interaction with c-myc promoter G-quadruplex, J. Med. Chem. 54 (2011)
[150] L. Ji, M. Arcinas, LM Boxer, The transcription factor, Nm23H2, binds to and activates the 5671–5679.
translocated c-myc allele in Burkitt's lymphoma, J. Biol. Chem. 270 (1995) 13392– [165] HY Liu, AC Chen, QK Yin, Z. Li, SM Huang, G. Du, JH He, LP Zan, SK Wang, YH Xu,
13398. JH Tan, TM Ou, D. Li, LQ Gu, ZS Huang, New dis ubstituted quindoline derivatives inhibiting
[151] TS Dexheimer, SS Carey, S. Zuohe, VM Gokhale, X. Hu, LB Murata, EM Maes, A. Burkitt's lymphoma cell proliferation by impeding c-MYC transcription, J. Med. Chem. 60
Weichsel, D. Sun, EJ Meuillet, WR Montfort, LH Hurley, NM23- H2 may play an (2017) 5438–5454.
indirect role in transcriptional activation of c-myc gene expression but does not cleave [166] J. Dai, M. Carver, LH Hurley, D. Yang, Solution structure of a 2:1 quindoline-c MYC G-
the nuclease hypersensitive element III(1), Mol. Cancer Ther. 8 (2009) 1363–1377. quadruplex: insights into G-quadruplex-interactive small molecule drug design, J. Am.
[152] M. Hildebrandt, ML Lacombe, S. Mesnildrey, M. Veron, A human NDP-kinase B Chem. Soc. 133 (2011) 17673–17680.
specifically binds single-stranded poly-pyrimidine sequences, Nucleic Acids Res. 23 [167] DY Zeng, GT Kuang, SK Wang, W. Peng, SL Lin, Q. Zhang, XX Su, MH Hu, H. Wang,
(1995) 3858–3864. JH Tan, ZS Huang, LQ Gu, TM Ou, Discovery of novel 11-triazole substituted
[153] RK Thakur, P. Kumar, K. Halder, A. Verma, A. Kar, JL Parent, R. Basundra, A. Kumar, benzofuro[3,2-b]quinolone derivatives as c-myc G-quadruplex specific stabilizers via
S. Chowdhury, Metastases suppressor NM23-H2 interaction with G quadruplex DNA click chemistry, J. Med. Chem. 60 (2017) 5407–5423.
within c-MYC promoter nuclease hypersensitive element induces c-MYC expression, [168] CL Grand, H. Han, RM Munoz, S. Weitman, DD Von Hoff, LH Hurley, DJ Bearss, The
Nucleic Acids Res. 37 (2009) 172–183. cationic porphyrin TMPyP4 down-regulates c-MYC and human telomerase reverse
[154] C. Shan, JW Yan, YQ Wang, T. Che, ZL Huang, AC Chen, PF Yao, JH Tan, D. Li, TM transcriptase expression and inhibits tumor growth in vivo, Mol. Cancer Ther. 1
Ou, LQ Gu, ZS Huang, Design, synthesis, and evaluation of isain digotone derivatives to (2002) 565–573.
downregulate c-myc transcription via disrupting the in teraction of NM23-H2 with G- [169] J. Seenisamy, EM Rezler, TJ Powell, D. Tye, V. Gokhale, CS Joshi, A. Siddiqui Jain, LH
quadruplex, J. Med. Chem. 60 (2017) 1292–1308. Hurley, The dynamic character of the G-quadruplex element in the c MYC promoter
[155] A. Fekete, E. Kenesi, E. Hunyadi-Gulyas, H. Durgo, B. Berko, ZA Dunai, PI Bauer, The and modification by TMPyP4, J. Am. Chem. Soc. 126 (2004) 8702–8709.
guanine-quadruplex structure in the human c-myc gene's promoter is converted into B- [170] J. Seenisamy, S. Bashyam, V. Gokhale, H. Vankayalapati, D. Sun, A. Siddiqui-Jain, N.
DNA form by the human poly(ADP-ribose)polymerase-1, PLoS One 7 (2012) e42690. Streiner, K. Shin-Ya, E. White, WD Wilson, LH Hurley, Design and synthesis of an
[156] L. Zandarashvili, MF Langelier, UK Velagapudi, MA Hancock, JD Steffen, R. Billur, ZM expanded porphyrin that has selectivity for the c-MYC G-quadruplex struc ture, J. Am.
Hannan, AJ Wicks, DB Krastev, SJ Pettitt, CJ Lord, TT Talele, JM Pascal, BE Black, Chem. Soc. 127 (2005) 2944–2959.
Structural basis for allosteric PARP-1 retention on DNA breaks, Science 368 (2020). [171] V. Gabelica, ES Baker, MP Teulade-Fichou, E. De Pauw, MT Bowers, Stabilization and
[157] MJ Suskiewicz, F. Zobel, TEH Ogden, P. Fontana, A. Ariza, JC Yang, K. Zhu, L. structure of telomeric and c-myc region intramolecular G quadruplexes: the role of
Bracken, WJ Hawthorne, D. Ahel, D. Neuhaus, I. Ahel, HPF1 completes the PARP central cations and small planar ligands, J. Am. Chem. Soc. 129 (2007) 895–904.
active site for DNA damage-induced ADP-ribosylation, Nature 579 (2020) 598–602. [172] MH Hu, YQ Wang, ZY Yu, LN Hu, TM Ou, SB Chen, ZS Huang, JH Tan, Discovery of a
[158] WL Kraus, Transcriptional control by PARP-1: chromatin modulation, enhancer binding, new four-leaf clover-like ligand as a potent c-MYC transcription inhibitor specifically
coregulation, and insulation, Curr. Opin. Biol Sel. 20 (2008) 294–302. [159] VA Soldatenkov, targeting the promoter G-quadruplex, J. Med. Chem. 61 (2018) 2447–2459.
S. Chasovskikh, VN Potaman, I. Trofimova, ME Smulson, A. Dritschilo, Transcriptional [173] A. Gluszynska, B. Juskowiak, M. Kuta-Siejkowska, M. Hoffmann, S. Haider,
repression by binding of poly(ADP-ribose) poly merase to promoter sequences, J. Biol. Chem. Carbazole ligands as c-myc G-quadruplex binders, Int. J. Biol. Macromol. 114
277 (2002) 665–670. (2018) 479–490.
[160] I. Lonskaya, VN Potaman, LS Shlyakhtenko, EA Oussatcheva, YL Lyubchenko, VA [174] T. Das, D. Panda, P. Saha, J. Dash, Small molecule driven stabilization of promoter G-
Soldatenkov, Regulation of poly(ADP-ribose) polymerase-1 by DNA structure specific quadruplexes and transcriptional regulation of c-MYC, Bioconjug. Chem. 29 (2018)
binding, J. Biol. Chem. 280 (2005) 17076–17083. 2636–2645.
[161] I. Mela, R. Kranaster, RM Henderson, S. Balasubramanian, JM Edwardson, [175] AW Schmidt, KR Reddy, HJ Knolker, Occurrence, biogenesis, and synthesis of
Demonstration of ligand decoration, and ligand-induced perturbation, of G biologically active carbazole alkaloids, Chem. Rev. 112 (2012) 3193–3328. [176] Y. Ma, TM
Ou, JQ Hou, YJ Lu, JH Tan, LQ Gu, ZS Huang, 9-N-Substituted
berberine derivatives: stabilization of G-quadruplex DNA and down-regulation of
oncogene c-myc, Bioorg. Med. Chem. 16 (2008) 7582–7591.
[177] Y. Ma, TM Ou, JH Tan, JQ Hou, SL Huang, LQ Gu, ZS Huang, Quinolino benzo-[5, 6]-
dihydroisoquindolium compounds derived from berberine: a new class of highly
selective ligands for G-quadruplex DNA in c-myc oncogene, Eur. J. Med. Chem. 46
(2011) 1906–1913.
[178] D. Peng, JH Tan, SB Chen, TM Ou, LQ Gu, ZS Huang, Bisaryldiketene de rivatives: a
new class of selective ligands for c-myc G-quadruplex DNA, Bioorg. Med. Chem. 18
(2010) 8235–8242.
[179] MM Islam, S. Fujii, S. Sato, T. Okauchi, S. Takenaka, A selective G-quadruplex DNA-
stabilizing ligand based on a cyclic naphthalene diimide derivative, Molecules 20
(2015) 10963–10979.
[180] DS Chan, H. Yang, MH Kwan, Z. Cheng, P. Lee, LP Bai, ZH Jiang, CY Wong, WF Fong,
CH Leung, DL Ma, Structure-based optimization of FDA-approved drug methylene blue
as a c-myc G-quadruplex DNA stabilizer, Biochimie 93 (2011) 1055–1064.
[181] J. Alzeer, NW Luedtke, pH-mediated fluorescence and G-quadruplex binding of amido
phthalocyanines, Biochemistry 49 (2010) 4339–4348.
[182] HM Lee, DS Chan, F. Yang, HY Lam, SC Yan, CM Che, DL Ma, CH Leung,
Identification of natural product fonsecin B as a stabilizing ligand of c-myc G
quadruplex DNA by high-throughput virtual screening, Chem. Komun. (Camb.) 46
(2010) 4680–4682.
[183] P. Wu, DL Ma, CH Leung, SC Yan, N. Zhu, R. Abagyan, CM Che, Stabilization of G-
quadruplex DNA with platinum(II) Schiff base complexes: luminescent probe and
down-regulation of c-myc oncogene expression, Chemistry (Easton) 15 (2009) 13008–
13021.
[184] ZF Chen, QP Qin, JL Qin, YC Liu, KB Huang, YL Li, T. Meng, GH Zhang, Y. Peng, XJ
Luo, H. Liang, Stabilization of G-quadruplex DNA, inhibition of tel omerase activity, and
tumor cell apoptosis by organoplatinum(II) complexes with oxoisoaporphine, J. Med.
Chem. 58 (2015) 2159–2179.
[185] C. Wei, L. Ren, N. Gao, Interactions of terpyridines and their Pt(II) complexes with G-
quadruplex DNAs and telomerase inhibition, Int. J. Biol. Macromol. 57 (2013) 1–8.
W. Wang, et al. BBA - Reviews on Cancer 1874 (2020) 188410

[186] P. Wang, CH Leung, DL Ma, SC Yan, CM Che, Structure-based design of [192] A. Tawani, A. Amanullah, A. Mishra, A. Kumar, Evidences for Piperine inhibiting cancer
platinum(II) complexes as c-myc oncogene down-regulators and luminescent by targeting human G-quadruplex DNA sequences, Sci. Rep. 6 (2016) 39239.
probes for G-quadruplex DNA, Chemistry (Easton) 16 (2010) 6900–6911. [193] A. Tawani, SK Mishra, A. Kumar, Structural insight for the recognition of G quadruplex
[187] KV Diveshkumar, S. Sakrikar, F. Rosu, S. Harikrishna, V. Gabelica, PI Pradeepkumar, structure at human c-myc promoter sequence by flavonoid Quercetin, Sci. Rep. 7
Specific stabilization of c-MYC and c-KIT G-quadruplex DNA structures by (2017) 3600.
indolylmethyleneindanone scaffolds, Biochemistry 55 (2016) 3571–3585. [194] RV Brown, FL Danford, V. Gokhale, LH Hurley, TA Brooks, Demonstration that drug-
[188] P. Murat, Y. Singh, E. Defrancq, Methods for investigating G-quadruplex DNA/ ligand targeted down-regulation of MYC in non-Hodgkins lymphoma is directly mediated
interactions, Chem. Soc. Rev. 40 (2011) 5293–5307. through the promoter G-quadruplex, J. Biol. Chem. 286 (2011) 41018–41027.
[189] YX Xiong, ZS Huang, JH Tan, Targeting G-quadruplex nucleic acids with [195] C. Weldon, JG Dacanay, V. Gokhale, PVL Boddupally, I. Behm-Ansmant, GA Burley, C.
heterocyclic alkaloids and their derivatives, Eur. J. Med. Chem. 97 (2015) 538– Branlant, LH Hurley, C. Dominguez, IC Eperon, Specific G quadruplex ligands
551. modulate the alternative splicing of Bcl-X, Nucleic Acids Res. 46 (2018) 886–896.
[190] S. Zhang, Y. Wu, W. Zhang, G-quadruplex structures and their interaction diversity with [196] JJ Montoya, MA Turnidge, DH Wai, AR Patel, DW Lee, V. Gokhale, LH Hurley, RJ
ligands, ChemMedChem 9 (2014) 899–911. Arceci, C. Wetmore, DO Azorsa, In vitro activity of a G-quad ruplex-stabilizing small
[191] X. Ji, H. Sun, H. Zhou, J. Xiang, Y. Tang, C. Zhao, The interaction of telomeric DNA and molecule that synergizes with Navitoclax to induce cyto toxicity in acute myeloid
C-myc22 G-quadruplex with 11 natural alkaloids, Nucleic Acid Ther. 22 (2012) 127– leukemia cells, BMC Cancer 19 (2019) 1251.
136. [197] D. Panda, P. Saha, T. Das, J. Dash, Target guided synthesis using DNA nano templates
for selectively assembling a G-quadruplex binding c-MYC inhibitor, Nat. Komun. 8 16
(2017) 16103. TC Jenkins, S. Neidle, LH Hurley, Inhibition of human telomerase by a G quadruplex-
[198] KM Felsenstein, LB Saunders, JK Simmons, E. Leon, DR Calabrese, S. Zhang, A. interactive compound, J. Med. Chem. 40 (1997) 2113–2116. [201] A. Randazzo, A. Galeone,
Michalowski, P. Gareiss, BA Mock, JS Schneekloth Jr., Small molecule mi croarrays V. Esposito, M. Varra, L. Mayol, Interaction of dis tamycin A and netropsin with quadruplex
enable the identification of a selective, quadruplex-binding inhibitor of MYC expression, and duplex structures: a comparative 1H-NMR study, Nucleosides Nucleotides Nucleic Acids
ACS Chem. Biol. 11 (2016) 139–148. 21 (2002) 535–545. [202] A. Ali, M. Kamra, S. Roy, K. Muniyappa, S. Bhattacharya, Novel
[199] R. Kumar, K. Chand, S. Bhowmik, RN Das, S. Bhattacharjee, M. Hedenstrom, E. oligopyrrole carboxamide based nickel(II) and palladium(II) salens, their targeting of human G
Chorell, Subtle structural alterations in G-quadruplex DNA regulate site speci ficity of quadruplex DNA, and selective cancer cell toxicity, Chem. Asian J. 11 (2016) 2542–2554.
fluorescence light-up probes, Nucleic Acids Res. 48 (2020) 1108–1119. [200] D. Sun, B. [203] D. Drygin, A. Siddiqui-Jain, S. O'Brien, M. Schwaebe, A. Lin, J. Bliesath, CB Ho, C.
Thompson, BE Cathers, M. Salazar, SM Kerwin, JO Trent, Proffitt, K. Trent, JP Whitten, JK Lim, D. Von Hoff, K. Anderes, WG Rice, Anticancer
activity of CX-3543: a direct inhibitor of rRNA biogenesis, Cancer Res. 69 (2009)
7653–7661.
[204] TA Brooks, LH Hurley, Targeting MYC expression through G-quadruplexes, Genes
Cancer 1 (2010) 641–649.
[205] YQ Wang, ZL Huang, SB Chen, CX Wang, C. Shan, QK Yin, TM Ou, D. Li, LQ Gu, JH
Tan, ZS Huang, Design, synthesis, and evaluation of new selective NM23-H2 binders as c-
MYC transcription inhibitors via disruption of the NM23- H2/G-quadruplex interaction, J. Med.
Chem. 60 (2017) 6924–6941.
[206] C. Bouvard, SM Lim, J. Ludka, N. Yazdani, AK Woods, AK Chatterjee, PG Schultz, S.
Zhu, Small molecule selectively suppresses MYC transcription in cancer cells, Proc. Natl.
Acad. Sci. USA 114 (2017) 3497–3502.
[207] D. Yang, K. Okamoto, Structural insights into G-quadruplexes: towards new an ticancer
drugs, Future Med. Chem. 2 (2010) 619–646.
[208] V. Le Joncour, P. Laakkonen, Seek & destroy, use of targeting peptides for cancer
detection and drug delivery, Bioorg. Med. Chem. 26 (2018) 2797–2806. [209] Y. De, Q. Chen,
AP Schmidt, GM Anderson, JM Wang, J. Wooters, JJ Oppenheim, O. Chertov, LL-37, the
neutrophil granule- and epithelial cell derived cathelicidin, utilizes formyl peptide receptor-like
1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and
T cells, J. Exp. Med. 192 (2000) 1069–1074.
[210] L. Sun, W. Wang, W. Xiao, H. Yang, The roles of cathelicidin LL-37 in in flammatory
bowel disease, Inflamm. Bowel Dis. 22 (2016) 1986–1991. [211] M. Marzano, AP Falanga,
D. Marasco, N. Borbone, S. D'Errico, G. Piccialli, GN Roviello, G. Oliviero, Evaluation of an
analogue of the marine epsilon-PLL peptide as a ligand of G-quadruplex DNA structures,
Mar. Drugs 18 (2020). [212] NK Lee, HD Paik, Status, antimicrobial mechanism, and
regulation of natural preservatives in livestock food systems, Korean J. Food Sci. Anim.
Resour. 36 (2016) 547–557.
[213] W. Bao, R. Liu, Y. Wang, F. Wang, G. Xia, H. Zhang, X. Li, H. Yin, B. Chen, PLGA PLL-
PEG-Tf-based targeted nanoparticles drug delivery system enhance antitumor efficacy
via intrinsic apoptosis pathway, Int. J. Nanomedicine 10 (2015) 557–566.
[214] J. Du, C. Tian, J. Ling, Y. Wang, R8-modified polysarcosine-b-polylysine poly peptide
to enhance circulation stability and gene delivery efficiency, J. Control. Release 213
(2015) e50–e51.

Anda mungkin juga menyukai