Skip to content
2000
image of The Expression of the LDLR, LDLRAP1, and PCSK9 Genes has Prognostic Significance in Triple-negative Breast Cancer

Abstract

Aims

The purpose of this study was to investigate the prognostic significance of cholesterol uptake genes in predicting the survival of breast cancer patients.

Background

Cholesterol plays a crucial role in the homeostasis of tumor cells. It is known that cholesterol levels can influence important parameters of the disease, such as sensitivity to therapy, progression, and metastasis of cancer. Previous studies suggest that breast cancer subtypes exhibit differences in metabolism.

Objective

The objectives of this study were to determine whether cholesterol uptake genes have prognostic significance for overall survival in breast cancer patients, evaluate if this prognostic significance varies between breast cancer subtypes, and identify differences in the expression of cholesterol uptake genes among these subtypes.

Methods

Data from mRNA sequencing of tumors from the Cancer Genome Atlas (TCGA) portal were analyzed. Tumors were classified into molecular subtypes, and the prognostic significance of cholesterol uptake gene expression levels was evaluated for each subtype. DESeq2 and Fisher's test were used to assess differences in gene expression.

Results

High expression levels of genes involved in de novo cholesterol synthesis were associated with poor prognosis for the Basal-like and Luminal A breast cancer subtypes. The prognostic significance of low-density lipoprotein receptor (LDLR), LDLR adapter protein 1 (LDLRAP1), and proprotein convertase subtilisin/kexin type 9 (PCSK9), which are responsible for exogenous cholesterol uptake, varied across subtypes. Specifically, low expression of LDLR was associated with a favorable prognosis for the luminal A (OR = 2.17; FDR = 0.0048) and luminal B (OR = 2.21; FDR = 0.015) subtypes but indicated poor prognosis in the basal-like subtype (OR = 0.48; FDR = 0.05). No genes were significant for prognosis prediction in the HER2-positive subtype. The HER2+ subtype exhibited higher expression of cholesterol uptake genes compared to the basal-like subtype based on the analysis of tumor mRNA sequencing (OR = 6.45, -value = 3.07E-05). This finding was also confirmed through the study of publicly available single-cell sequencing data (OR = 40.3, -value = 2.19e-07), which may contribute to the differences in their prognostic significance.

Conclusion

The prognostic significance of cholesterol uptake gene expression varies among breast cancer subtypes. Precise fitting of biomarkers into breast cancer subtypes may aid in more accurate patient stratification and improve treatment approaches.

Loading

Article metrics loading...

/content/journals/cmc/10.2174/0109298673291217241219055416
2025-01-20
2025-04-25
Loading full text...

Full text loading...

References

  1. Tran K.B. Lang J.J. Compton K. Xu R. Acheson A.R. Henrikson H.J. Kocarnik J.M. Penberthy L. Aali A. Abbas Q. Abbasi B. Kangevari A.M. Kangevari A.Z. Abbastabar H. Abdelmasseh M. Abd-Elsalam S. Abdelwahab A.A. Abdoli G. Abdulkadir H.A. Abedi A. Abegaz K.H. Abidi H. Aboagye R.G. Abolhassani H. Absalan A. Abtew Y.D. Ali A.H. Gharbieh A.E. Achappa B. Acuna J.M. Addison D. Addo I.Y. Adegboye O.A. Adesina M.A. Adnan M. Adnani Q.E.S. Advani S.M. Afrin S. Afzal M.S. Aggarwal M. Ahinkorah B.O. Ahmad A.R. Ahmad R. Ahmad S. Ahmad S. Ahmadi S. Ahmed H. Ahmed L.A. Ahmed M.B. Rashid A.T. Aiman W. Ajami M. Akalu G.T. Khiavi A.M. Aklilu A. Akonde M. Akunna C.J. Al Hamad H. Alahdab F. Alanezi F.M. Alanzi T.M. Alessy S.A. Algammal A.M. Al-Hanawi M.K. Alhassan R.K. Ali B.A. Ali L. Ali S.S. Alimohamadi Y. Alipour V. Aljunid S.M. Alkhayyat M. Al-Maweri S.A.A. Almustanyir S. Alonso N. Alqalyoobi S. Al-Raddadi R.M. Al-Rifai R.H.H. Al-Sabah S.K. Al-Tammemi A.B. Altawalah H. Guzman A.N. Amare F. Ameyaw E.K. Dehkordi A.J.J. Iranaq A.M.H. Amu H. Amusa G.A. Ancuceanu R. Anderson J.A. Animut Y.A. Anoushiravani A. Anoushirvani A.A. Moghaddam A.A. Ansha M.G. Antony B. Antwi M.H. Anwar S.L. Anwer R. Anyasodor A.E. Arabloo J. Zozani A.M. Aremu O. Argaw A.M. Ariffin H. Aripov T. Arshad M. Artaman A. Arulappan J. Aruleba R.T. Aryannejad A. Asaad M. Asemahagn M.A. Asemi Z. Jafarabadi A.M. Ashraf T. Assadi R. Athar M. Athari S.S. Atout M.M.W. Attia S. Aujayeb A. Ausloos M. Burgos A.L. Awedew A.F. Awoke M.A. Awoke T. Quintanilla A.B.P. Ayana T.M. Ayen S.S. Azadi D. Azadnajafabad S. Aghdash A.S. Azanaw M.M. Khyavy A.M. Jafari A.A. Azizi H. Azzam A.Y.Y. Babajani A. Badar M. Badiye A.D. Baghcheghi N. Bagheri N. Bagherieh S. Bahadory S. Baig A.A. Baker J.L. Bakhtiari A. Bakshi R.K. Banach M. Banerjee I. Bardhan M. Adesi B.F. Barra F. Barrow A. Bashir N.Z. Bashiri A. Basu S. Batiha A-M.M. Begum A. Bekele A.B. Belay A.S. Belete M.A. Belgaumi U.I. Bell A.W. Belo L. Benzian H. Berhie A.Y. Bermudez A.N.C. Bernabe E. Bhagavathula A.S. Bhala N. Bhandari B.B. Bhardwaj N. Bhardwaj P. Bhattacharyya K. Bhojaraja V.S. Bhuyan S.S. Bibi S. Bilchut A.H. Bintoro B.S. Biondi A. Birega M.G.B. Birhan H.E. Bjørge T. Blyuss O. Bodicha B.B.A. Bolla S.R. Boloor A. Bosetti C. Braithwaite D. Brauer M. Brenner H. Briko A.N. Briko N.I. Buchanan C.M. Bulamu N.B. Teixeira B.M.T. Butt M.H. Butt N.S. Butt Z.A. Santos C.F.L. Cámera L.A. Cao C. Cao Y. Carreras G. Carvalho M. Cembranel F. Cerin E. Chakraborty P.A. Charalampous P. Chattu V.K. Chimed-Ochir O. Caceres C.J.L. Cho D.Y. Cho W.C.S. Christopher D.J. Chu D-T. Chukwu I.S. Cohen A.J. Conde J. Cortés S. Costa V.M. Martins C.N. Culbreth G.T. Dadras O. Dagnaw F.T. Dahlawi S.M.A. Dai X. Dandona L. Dandona R. Daneshpajouhnejad P. Danielewicz A. Dao A.T.M. Soltani D.C.R. Darwesh A.M. Das S. Davitoiu D.V. Esmaeili D.E. Hoz D.L.F.P. Debela S.A. Dehghan A. Demisse B. Demisse F.W. Gutiérrez D.E. Derakhshani A. Derbew Molla M. Dereje D. Deribe K.S. Desai R. Desalegn M.D. Dessalegn F.N. Dessalegni S.A.A. Dessie G. Desta A.A. Dewan S.M.R. Dharmaratne S.D. Dhimal M. Dianatinasab M. Diao N. Diaz D. Digesa L.E. Dixit S.G. Doaei S. Doan L.P. Doku P.N. Dongarwar D. dos Santos W.M. Driscoll T.R. Dsouza H.L. Durojaiye O.C. Edalati S. Eghbalian F. Ehsani-Chimeh E. Eini E. Ekholuenetale M. Ekundayo T.C. Ekwueme D.U. El Tantawi M. Elbahnasawy M.A. Elbarazi I. Elghazaly H. Elhadi M. El-Huneidi W. Emamian M.H. Engelbert Bain L. Enyew D.B. Erkhembayar R. Eshetu T. Eshrati B. Eskandarieh S. Espinosa-Montero J. Etaee F. Etemadimanesh A. Eyayu T. Ezeonwumelu I.J. Ezzikouri S. Fagbamigbe A.F. Fahimi S. Fakhradiyev I.R. Faraon E.J.A. Fares J. Farmany A. Farooque U. Farrokhpour H. Fasanmi A.O. Fatehizadeh A. Fatima W. Fattahi H. Fekadu G. Feleke B.E. Ferrari A.A. Ferrero S. Ferro Desideri L. Filip I. Fischer F. Foroumadi R. Foroutan M. Fukumoto T. Gaal P.A. Gad M.M. Gadanya M.A. Gaipov A. Galehdar N. Gallus S. Garg T. Gaspar Fonseca M. Gebremariam Y.H. Gebremeskel T.G. Gebremichael M.A. Geda Y.F. Gela Y.Y. Gemeda B.N.B. Getachew M. Getachew M.E. Ghaffari K. Ghafourifard M. Ghamari S-H. Ghasemi Nour M. Ghassemi F. Ghimire A. Ghith N. Gholamalizadeh M. Gholizadeh Navashenaq J. Ghozy S. Gilani S.A. Gill P.S. Ginindza T.G. Gizaw A.T.T. Glasbey J.C. Godos J. Goel A. Golechha M. Goleij P. Golinelli D. Golitaleb M. Gorini G. Goulart B.N.G. Grosso G. Guadie H.A. Gubari M.I.M. Gudayu T.W. Guerra M.R. Gunawardane D.A. Gupta B. Gupta S. Gupta V.B. Gupta V.K. Gurara M.K. Guta A. Habibzadeh P. Haddadi Avval A. Hafezi-Nejad N. Hajj Ali A. Haj-Mirzaian A. Halboub E.S. Halimi A. Halwani R. Hamadeh R.R. Hameed S. Hamidi S. Hanif A. Hariri S. Harlianto N.I. Haro J.M. Hartono R.K. Hasaballah A.I. Hasan S.M.M. Hasani H. Hashemi S.M. Hassan A.M. Hassanipour S. Hayat K. Heidari G. Heidari M. Heidarymeybodi Z. Herrera-Serna B.Y. Herteliu C. Hezam K. Hiraike Y. Hlongwa M.M. Holla R. Holm M. Horita N. Hoseini M. Hossain M.M. Hossain M.B.H. Hosseini M-S. Hosseinzadeh A. Hosseinzadeh M. Hostiuc M. Hostiuc S. Househ M. Huang J. Hugo F.N. Humayun A. Hussain S. Hussein N.R. Hwang B-F. Ibitoye S.E. Iftikhar P.M. Ikuta K.S. Ilesanmi O.S. Ilic I.M. Ilic M.D. Immurana M. Innos K. Iranpour P. Irham L.M. Islam M.S. Islam R.M. Islami F. Ismail N.E. Isola G. Iwagami M. J L.M. Jaiswal A. Jakovljevic M. Jalili M. Jalilian S. Jamshidi E. Jang S-I. Jani C.T. Javaheri T. Jayarajah U.U. Jayaram S. Jazayeri S.B. Jebai R. Jemal B. Jeong W. Jha R.P. Jindal H.A. John-Akinola Y.O. Jonas J.B. Joo T. Joseph N. Joukar F. Jozwiak J.J. Jürisson M. Kabir A. Kacimi S.E.O. Kadashetti V. Kahe F. Kakodkar P.V. Kalankesh L.R. Kalankesh L.R. Kalhor R. Kamal V.K. Kamangar F. Kamath A. Kanchan T. Kandaswamy E. Kandel H. Kang H.J. Kanno G.G. Kapoor N. Kar S.S. Karanth S.D. Karaye I.M. Karch A. Karimi A. Kassa B.G. Katoto P.D.M.C. Kauppila J.H. Kaur H. Kebede A.G. Keikavoosi-Arani L. Kejela G.G. Kemp Bohan P.M. Keramati M. Keykhaei M. Khajuria H. Khan A. Khan A.A.K. Khan E.A. Khan G. Khan M.N. Khan M.A.B. Khanali J. Khatab K. Khatatbeh M.M. Khatib M.N. Khayamzadeh M. Khayat Kashani H.R. Khazeei Tabari M.A. Khezeli M. Khodadost M. Kim M.S. Kim Y.J. Kisa A. Kisa S. Klugar M. Klugarová J. Kolahi A-A. Kolkhir P. Kompani F. Koul P.A. Koulmane Laxminarayana S.L. Koyanagi A. Krishan K. Krishnamoorthy Y. Kucuk Bicer B. Kugbey N. Kulimbet M. Kumar A. Kumar G.A. Kumar N. Kurmi O.P. Kuttikkattu A. La Vecchia C. Lahiri A. Lal D.K. Lám J. Lan Q. Landires I. Larijani B. Lasrado S. Lau J. Lauriola P. Ledda C. Lee S. Lee S.W.H. Lee W-C. Lee Y.Y. Lee Y.H. Legesse S.M. Leigh J. Leong E. Li M-C. Lim S.S. Liu G. Liu J. Lo C-H. Lohiya A. Lopukhov P.D. Lorenzovici L. Lotfi M. Loureiro J.A. Lunevicius R. Madadizadeh F. Mafi A.R. Magdeldin S. Mahjoub S. Mahmoodpoor A. Mahmoudi M. Mahmoudimanesh M. Mahumud R.A. Majeed A. Majidpoor J. Makki A. Makris K.C. Malakan Rad E. Malekpour M-R. Malekzadeh R. Malik A.A. Mallhi T.H. Mallya S.D. Mamun M.A. Manda A.L. Mansour-Ghanaei F. Mansouri B. Mansournia M.A. Mantovani L.G. Martini S. Martorell M. Masoudi S. Masoumi S.Z. Matei C.N. Mathews E. Mathur M.R. Mathur V. McKee M. Meena J.K. Mehmood K. Mehrabi Nasab E. Mehrotra R. Melese A. Mendoza W. Menezes R.G. Mengesha S.I.D. Mensah L.G. Mentis A-F.A. Mera-Mamián A.Y.M. Meretoja T.J. Merid M.W. Mersha A.G. Meselu B.T. Meshkat M. Mestrovic T. Miao Jonasson J. Miazgowski T. Michalek I.M. Mijena G.F.W. Miller T.R. Mir S.A. Mirinezhad S.K. Mirmoeeni S. Mirza-Aghazadeh-Attari M. Mirzaei H. Mirzaei H.R. Misganaw A.S. Misra S. Mohammad K.A. Mohammadi E. Mohammadi M. Mohammadian-Hafshejani A. Mohammadpourhodki R. Mohammed A. Mohammed S. Mohan S. Mohseni M. Moka N. Mokdad A.H. Molassiotis A. Molokhia M. Momenzadeh K. Momtazmanesh S. Monasta L. Mons U. Montasir A.A. Montazeri F. Montero A. Moosavi M.A. Moradi A. Moradi Y. Moradi Sarabi M. Moraga P. Morawska L. Morrison S.D. Morze J. Mosapour A. Mostafavi E. Mousavi S.M. Mousavi Isfahani H. Mousavi Khaneghah A. Mpundu-Kaambwa C. Mubarik S. Mulita F. Munblit D. Munro S.B. Murillo-Zamora E. Musa J. Nabhan A.F. Nagarajan A.J. Nagaraju S.P. Nagel G. Naghipour M. Naimzada M.D. Nair T.S. Naqvi A.A. Narasimha Swamy S. Narayana A.I. Nassereldine H. Natto Z.S. Nayak B.P. Ndejjo R. Nduaguba S.O. Negash W.W. Nejadghaderi S.A. Nejati K. Neupane Kandel S. Nguyen H.V.N. Niazi R.K. Noor N.M. Noori M. Noroozi N. Nouraei H. Nowroozi A. Nuñez-Samudio V. Nzoputam C.I. Nzoputam O.J. Oancea B. Odukoya O.O. Oghenetega O.B. Ogunsakin R.E. Oguntade A.S. Oh I-H. Okati-Aliabad H. Okekunle A.P. Olagunju A.T. Olagunju T.O. Olakunde B.O. Olufadewa I.I. Omer E. Omonisi A.E.E. Ong S. Onwujekwe O.E. Orru H. Otstavnov S.S. Oulhaj A. Oumer B. Owopetu O.F. Oyinloye B.E. P A M. Padron-Monedero A. Padubidri J.R. Pakbin B. Pakshir K. Pakzad R. Palicz T. Pana A. Pandey A. Pandey A. Pant S. Pardhan S. Park E-C. Park E-K. Park S. Patel J. Pati S. Paudel R. Paudel U. Paun M. Pazoki Toroudi H. Peng M. Pereira J. Pereira R.B. Perna S. Perumalsamy N. Pestell R.G. Pezzani R. Piccinelli C. Pillay J.D. Piracha Z.Z. Pischon T. Postma M.J. Pourabhari Langroudi A. Pourshams A. Pourtaheri N. Prashant A. Qadir M.M.F. Quazi Syed Z. Rabiee M. Rabiee N. Radfar A. Radhakrishnan R.A. Radhakrishnan V. Raeisi M. Rafiee A. Rafiei A. Raheem N. Rahim F. Rahman M.O. Rahman M. Rahman M.A. Rahmani A.M. Rahmani S. Rahmanian V. Rajai N. Rajesh A. Ram P. Ramezanzadeh K. Rana J. Ranabhat K. Ranasinghe P. Rao C.R. Rao S.J. Rashedi S. Rashidi A. Rashidi M. Rashidi M-M. Ratan Z.A. Rawaf D.L. Rawaf S. Rawal L. Rawassizadeh R. Razeghinia M.S. Rehman A.U. Rehman I. Reitsma M.B. Renzaho A.M.N. Rezaei M. Rezaei N. Rezaei N. Rezaei N. Rezaei S. Rezaeian M. Rezapour A. Riad A. Rikhtegar R. Rios-Blancas M. Roberts T.J. Rohloff P. Romero-Rodríguez E. Roshandel G. Rwegerera G.M. S M. Saber-Ayad M.M. Saberzadeh-Ardestani B. Sabour S. Saddik B. Sadeghi E. Saeb M.R. Saeed U. Safaei M. Safary A. Sahebazzamani M. Sahebkar A. Sahoo H. Sajid M.R. Salari H. Salehi S. Salem M.R. Salimzadeh H. Samodra Y.L. Samy A.M. Sanabria J. Sankararaman S. Sanmarchi F. Santric-Milicevic M.M. Saqib M.A.N. Sarveazad A. Sarvi F. Sathian B. Satpathy M. Sayegh N. Schneider I.J.C. Schwarzinger M. Šekerija M. Senthilkumaran S. Sepanlou S.G. Seylani A. Seyoum K. Sha F. Shafaat O. Shah P.A. Shahabi S. Shahid I. Shahrbaf M.A. Shahsavari H.R. Shaikh M.A. Shaka M.F. Shaker E. Shannawaz M. Sharew M.M.S. Sharifi A. Sharifi-Rad J. Sharma P. Shashamo B.B. Sheikh A. Sheikh M. Sheikhbahaei S. Sheikhi R.A. Sheikhy A. Shepherd P.R. Shetty A. Shetty J.K. Shetty R.S. Shibuya K. Shirkoohi R. Shirzad-Aski H. Shivakumar K.M. Shivalli S. Shivarov V. Shobeiri P. Shokri Varniab Z. Shorofi S.A. Shrestha S. Sibhat M.M. Siddappa Malleshappa S.K. Sidemo N.B. Silva D.A.S. Silva L.M.L.R. Silva Julian G. Silvestris N. Simegn W. Singh A.D. Singh A. Singh G. Singh H. Singh J.A. Singh J.K. Singh P. Singh S. Sinha D.N. Sinke A.H. Siraj M.S. Sitas F. Siwal S.S. Skryabin V.Y. Skryabina A.A. Socea B. Soeberg M.J. Sofi-Mahmudi A. Solomon Y. Soltani-Zangbar M.S. Song S. Song Y. Sorensen R.J.D. Soshnikov S. Sotoudeh H. Sowe A. Sufiyan M.B. Suk R. Suleman M. Suliankatchi Abdulkader R. Sultana S. Sur D. Szócska M. Tabaeian S.P. Tabarés-Seisdedos R. Tabatabaei S.M. Tabuchi T. Tadbiri H. Taheri E. Taheri M. Taheri Soodejani M. Takahashi K. Talaat I.M. Tampa M. Tan K-K. Tat N.Y. Tat V.Y. Tavakoli A. Tavakoli A. Tehrani-Banihashemi A. Tekalegn Y. Tesfay F.H. Thapar R. Thavamani A. Thoguluva Chandrasekar V. Thomas N. Thomas N.K. Ticoalu J.H.V. Tiyuri A. Tollosa D.N. Topor-Madry R. Touvier M. Tovani-Palone M.R. Traini E. Tran M.T.N. Tripathy J.P. Ukke G.G. Ullah I. Ullah S. Ullah S. Unnikrishnan B. Vacante M. Vaezi M. Valadan Tahbaz S. Valdez P.R. Vardavas C. Varthya S.B. Vaziri S. Velazquez D.Z. Veroux M. Villeneuve P.J. Violante F.S. Vladimirov S.K. Vlassov V. Vo B. Vu L.G. Wadood A.W. Waheed Y. Walde M.T. Wamai R.G. Wang C. Wang F. Wang N. Wang Y. Ward P. Waris A. Westerman R. Wickramasinghe N.D. Woldemariam M. Woldu B. Xiao H. Xu S. Xu X. Yadav L. Jabbari Y.S.H. Yang L. Yazdanpanah F. Yeshaw Y. Yismaw Y. Yonemoto N. Younis M.Z. Yousefi Z. Yousefian F. Yu C. Yu Y. Yunusa I. Zahir M. Zaki N. Zaman B.A. Zangiabadian M. Zare F. Zare I. Zareshahrabadi Z. Zarrintan A. Zastrozhin M.S. Zeineddine M.A. Zhang D. Zhang J. Zhang Y. Zhang Z-J. Zhou L. Zodpey S. Zoladl M. Vos T. Hay S.I. Force L.M. Murray C.J.L. The global burden of cancer attributable to risk factors, 2010–19: A systematic analysis for the global burden of disease study 2019. Lancet 2022 400 10352 563 591 10.1016/S0140‑6736(22)01438‑6 35988567
    [Google Scholar]
  2. Azamjah N. Zadeh S.Y. Zayeri F. Global trend of breast cancer mortality rate: A 25-year study. Asian Pacific J Cancer prevent. Asian Pac. J. Cancer Prev. 2019 20 7 2015 2020 10.31557/APJCP.2019.20.7.2015 31350959
    [Google Scholar]
  3. Kocarnik J.M. Compton K. Dean F.E. Fu W. Gaw B.L. Harvey J.D. Henrikson H.J. Lu D. Pennini A. Xu R. Ababneh E. Kangevari A.M. Abbastabar H. Abd-Elsalam S.M. Abdoli A. Abedi A. Abidi H. Abolhassani H. Adedeji I.A. Adnani Q.E.S. Advani S.M. Afzal M.S. Aghaali M. Ahinkorah B.O. Ahmad S. Ahmad T. Ahmadi A. Ahmadi S. Rashid A.T. Salih A.Y. Akalu G.T. Aklilu A. Akram T. Akunna C.J. Al Hamad H. Alahdab F. Al-Aly Z. Ali S. Alimohamadi Y. Alipour V. Aljunid S.M. Alkhayyat M. Almasi-Hashiani A. Almasri N.A. Al-Maweri S.A.A. Almustanyir S. Alonso N. Alvis-Guzman N. Amu H. Anbesu E.W. Ancuceanu R. Ansari F. Ansari-Moghaddam A. Antwi M.H. Anvari D. Anyasodor A.E. Aqeel M. Arabloo J. Arab-Zozani M. Aremu O. Ariffin H. Aripov T. Arshad M. Artaman A. Arulappan J. Asemi Z. Asghari Jafarabadi M. Ashraf T. Atorkey P. Aujayeb A. Ausloos M. Awedew A.F. Ayala Quintanilla B.P. Ayenew T. Azab M.A. Azadnajafabad S. Azari Jafari A. Azarian G. Azzam A.Y. Badiye A.D. Bahadory S. Baig A.A. Baker J.L. Balakrishnan S. Banach M. Bärnighausen T.W. Barone-Adesi F. Barra F. Barrow A. Behzadifar M. Belgaumi U.I. Bezabhe W.M.M. Bezabih Y.M. Bhagat D.S. Bhagavathula A.S. Bhardwaj N. Bhardwaj P. Bhaskar S. Bhattacharyya K. Bhojaraja V.S. Bibi S. Bijani A. Biondi A. Bisignano C. Bjørge T. Bleyer A. Blyuss O. Bolarinwa O.A. Bolla S.R. Braithwaite D. Brar A. Brenner H. Bustamante-Teixeira M.T. Butt N.S. Butt Z.A. Caetano dos Santos F.L. Cao Y. Carreras G. Catalá-López F. Cembranel F. Cerin E. Cernigliaro A. Chakinala R.C. Chattu S.K. Chattu V.K. Chaturvedi P. Chimed-Ochir O. Cho D.Y. Christopher D.J. Chu D.T. Chung M.T. Conde J. Cortés S. Cortesi P.A. Costa V.M. Cunha A.R. Dadras O. Dagnew A.B. Dahlawi S.M.A. Dai X. Dandona L. Dandona R. Darwesh A.M. das Neves J. De la Hoz F.P. Demis A.B. Denova-Gutiérrez E. Dhamnetiya D. Dhimal M.L. Dhimal M. Dianatinasab M. Diaz D. Djalalinia S. Do H.P. Doaei S. Dorostkar F. dos Santos Figueiredo F.W. Driscoll T.R. Ebrahimi H. Eftekharzadeh S. El Tantawi M. El-Abid H. Elbarazi I. Elhabashy H.R. Elhadi M. El-Jaafary S.I. Eshrati B. Eskandarieh S. Esmaeilzadeh F. Etemadi A. Ezzikouri S. Faisaluddin M. Faraon E.J.A. Fares J. Farzadfar F. Feroze A.H. Ferrero S. Ferro Desideri L. Filip I. Fischer F. Fisher J.L. Foroutan M. Fukumoto T. Gaal P.A. Gad M.M. Gadanya M.A. Gallus S. Gaspar Fonseca M. Getachew Obsa A. Ghafourifard M. Ghashghaee A. Ghith N. Gholamalizadeh M. Gilani S.A. Ginindza T.G. Gizaw A.T.T. Glasbey J.C. Golechha M. Goleij P. Gomez R.S. Gopalani S.V. Gorini G. Goudarzi H. Grosso G. Gubari M.I.M. Guerra M.R. Guha A. Gunasekera D.S. Gupta B. Gupta V.B. Gupta V.K. Gutiérrez R.A. Hafezi-Nejad N. Haider M.R. Haj-Mirzaian A. Halwani R. Hamadeh R.R. Hameed S. Hamidi S. Hanif A. Haque S. Harlianto N.I. Haro J.M. Hasaballah A.I. Hassanipour S. Hay R.J. Hay S.I. Hayat K. Heidari G. Heidari M. Herrera-Serna B.Y. Herteliu C. Hezam K. Holla R. Hossain M.M. Hossain M.B.H. Hosseini M.S. Hosseini M. Hosseinzadeh M. Hostiuc M. Hostiuc S. Househ M. Hsairi M. Huang J. Hugo F.N. Hussain R. Hussein N.R. Hwang B.F. Iavicoli I. Ibitoye S.E. Ida F. Ikuta K.S. Ilesanmi O.S. Ilic I.M. Ilic M.D. Irham L.M. Islam J.Y. Islam R.M. Islam S.M.S. Ismail N.E. Isola G. Iwagami M. Jacob L. Jain V. Jakovljevic M.B. Javaheri T. Jayaram S. Jazayeri S.B. Jha R.P. Jonas J.B. Joo T. Joseph N. Joukar F. Jürisson M. Kabir A. Kahrizi D. Kalankesh L.R. Kalhor R. Kaliyadan F. Kalkonde Y. Kamath A. Kameran Al-Salihi N. Kandel H. Kapoor N. Karch A. Kasa A.S. Katikireddi S.V. Kauppila J.H. Kavetskyy T. Kebede S.A. Keshavarz P. Keykhaei M. Khader Y.S. Khalilov R. Khan G. Khan M. Khan M.N. Khan M.A.B. Khang Y.H. Khater A.M. Khayamzadeh M. Kim G.R. Kim Y.J. Kisa A. Kisa S. Kissimova-Skarbek K. Kopec J.A. Koteeswaran R. Koul P.A. Koulmane Laxminarayana S.L. Koyanagi A. Kucuk Bicer B. Kugbey N. Kumar G.A. Kumar N. Kumar N. Kurmi O.P. Kutluk T. La Vecchia C. Lami F.H. Landires I. Lauriola P. Lee S. Lee S.W.H. Lee W.C. Lee Y.H. Leigh J. Leong E. Li J. Li M.C. Liu X. Loureiro J.A. Lunevicius R. Magdy Abd El Razek M. Majeed A. Makki A. Male S. Malik A.A. Mansournia M.A. Martini S. Masoumi S.Z. Mathur P. McKee M. Mehrotra R. Mendoza W. Menezes R.G. Mengesha E.W. Mesregah M.K. Mestrovic T. Miao Jonasson J. Miazgowski B. Miazgowski T. Michalek I.M. Miller T.R. Mirzaei H. Mirzaei H.R. Misra S. Mithra P. Moghadaszadeh M. Mohammad K.A. Mohammad Y. Mohammadi M. Mohammadi S.M. Mohammadian-Hafshejani A. Mohammed S. Moka N. Mokdad A.H. Molokhia M. Monasta L. Moni M.A. Moosavi M.A. Moradi Y. Moraga P. Morgado-da-Costa J. Morrison S.D. Mosapour A. Mubarik S. Mwanri L. Nagarajan A.J. Nagaraju S.P. Nagata C. Naimzada M.D. Nangia V. Naqvi A.A. Narasimha Swamy S. Ndejjo R. Nduaguba S.O. Negoi I. Negru S.M. Neupane Kandel S. Nguyen C.T. Nguyen H.L.T. Niazi R.K. Nnaji C.A. Noor N.M. Nuñez-Samudio V. Nzoputam C.I. Oancea B. Ochir C. Odukoya O.O. Ogbo F.A. Olagunju A.T. Olakunde B.O. Omar E. Omar Bali A. Omonisi A.E.E. Ong S. Onwujekwe O.E. Orru H. Ortega-Altamirano D.V. Otstavnov N. Otstavnov S.S. Owolabi M.O. P A M. Padubidri J.R. Pakshir K. Pana A. Panagiotakos D. Panda-Jonas S. Pardhan S. Park E.C. Park E.K. Pashazadeh Kan F. Patel H.K. Patel J.R. Pati S. Pattanshetty S.M. Paudel U. Pereira D.M. Pereira R.B. Perianayagam A. Pillay J.D. Pirouzpanah S. Pishgar F. Podder I. Postma M.J. Pourjafar H. Prashant A. Preotescu L. Rabiee M. Rabiee N. Radfar A. Radhakrishnan R.A. Radhakrishnan V. Rafiee A. Rahim F. Rahimzadeh S. Rahman M. Rahman M.A. Rahmani A.M. Rajai N. Rajesh A. Rakovac I. Ram P. Ramezanzadeh K. Ranabhat K. Ranasinghe P. Rao C.R. Rao S.J. Rawassizadeh R. Razeghinia M.S. Renzaho A.M.N. Rezaei N. Rezaei N. Rezapour A. Roberts T.J. Rodriguez J.A.B. Rohloff P. Romoli M. Ronfani L. Roshandel G. Rwegerera G.M. S M. Sabour S. Saddik B. Saeed U. Sahebkar A. Sahoo H. Salehi S. Salem M.R. Salimzadeh H. Samaei M. Samy A.M. Sanabria J. Sankararaman S. Santric-Milicevic M.M. Sardiwalla Y. Sarveazad A. Sathian B. Sawhney M. Saylan M. Schneider I.J.C. Sekerija M. Seylani A. Shafaat O. Shaghaghi Z. Shaikh M.A. Shamsoddin E. Shannawaz M. Sharma R. Sheikh A. Sheikhbahaei S. Shetty A. Shetty J.K. Shetty P.H. Shibuya K. Shirkoohi R. Shivakumar K.M. Shivarov V. Siabani S. Siddappa Malleshappa S.K. Silva D.A.S. Singh J.A. Sintayehu Y. Skryabin V.Y. Skryabina A.A. Soeberg M.J. Sofi-Mahmudi A. Sotoudeh H. Steiropoulos P. Straif K. Subedi R. Sufiyan M.B. Sultan I. Sultana S. Sur D. Szerencsés V. Szócska M. Tabarés-Seisdedos R. Tabuchi T. Tadbiri H. Taherkhani A. Takahashi K. Talaat I.M. Tan K.K. Tat V.Y. Tedla B.A.A. Tefera Y.G. Tehrani-Banihashemi A. Temsah M.H. Tesfay F.H. Tessema G.A. Thapar R. Thavamani A. Thoguluva Chandrasekar V. Thomas N. Tohidinik H.R. Touvier M. Tovani-Palone M.R. Traini E. Tran B.X. Tran K.B. Tran M.T.N. Tripathy J.P. Tusa B.S. Ullah I. Ullah S. Umapathi K.K. Unnikrishnan B. Upadhyay E. Vacante M. Vaezi M. Valadan Tahbaz S. Velazquez D.Z. Veroux M. Violante F.S. Vlassov V. Vo B. Volovici V. Vu G.T. Waheed Y. Wamai R.G. Ward P. Wen Y.F. Westerman R. Winkler A.S. Yadav L. Yahyazadeh Jabbari S.H. Yang L. Yaya S. Yazie T.S.Y. Yeshaw Y. Yonemoto N. Younis M.Z. Yousefi Z. Yu C. Yuce D. Yunusa I. Zadnik V. Zare F. Zastrozhin M.S. Zastrozhina A. Zhang J. Zhong C. Zhou L. Zhu C. Ziapour A. Zimmermann I.R. Fitzmaurice C. Murray C.J.L. Force L.M. Global burden of disease 2019 cancer collaboration. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: A systematic analysis for the global burden of disease study 2019. JAMA Oncol. 2022 8 3 420 444 10.1001/jamaoncol.2021.6987 34967848
    [Google Scholar]
  4. Azadnajafabad S. Moghaddam S.S. Keykhaei M. Shobeiri P. Rezaei N. Ghasemi E. Mohammadi E. Ahmadi N. Ghamari A. Shahin S. Rezaei N. Aghili M. Kaviani A. Larijani B. Farzadfar F. Expansion of the quality of care index on breast cancer and its risk factors using the global burden of disease study 2019. Cancer Med. 2023 12 2 1729 1743 10.1002/cam4.4951 35770711
    [Google Scholar]
  5. Sung H. Ferlay J. Siegel R.L. Laversanne M. Soerjomataram I. Jemal A. Bray F. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021 71 3 209 249 10.3322/caac.21660 33538338
    [Google Scholar]
  6. Hauner D. Hauner H. Metabolic syndrome and breast cancer: Is there a link? Breast Care 2014 9 4 277 281 10.1159/000365951 25404888
    [Google Scholar]
  7. Karadag M.K. Akbulut M. Low HDL levels as the most common metabolic syndrome risk factor in heart failure. Int. Heart J. 2009 50 5 571 580 10.1536/ihj.50.571 19809206
    [Google Scholar]
  8. Denisenko Yulia K. Lipid-induced mechanisms of metabolic syndrome. J Obes. 2020 2020 5762395 10.1155/2020/5762395
    [Google Scholar]
  9. De Santi M. Annibalini G. Marano G. Biganzoli G. Venturelli E. Pellegrini M. Lucertini F. Brandi G. Biganzoli E. Barbieri E. Villarini A. Association between metabolic syndrome, insulin resistance, and IGF-1 in breast cancer survivors of DIANA-5 study. J. Cancer Res. Clin. Oncol. 2023 149 11 8639 8648 10.1007/s00432‑023‑04755‑6 37106164
    [Google Scholar]
  10. Nussbaumerova B. Rosolova H. Obesity and dyslipidemia. Curr. Atheroscler. Rep. 2023 25 12 947 955 10.1007/s11883‑023‑01167‑2 37979064
    [Google Scholar]
  11. Jha B.K. Sherpa M.L. Imran M. Mohammed Y. Jha L.A. Paudel K.R. Jha S.K. Progress in understanding metabolic syndrome and knowledge of its complex pathophysiology. Diabetology 2023 4 2 134 159 10.3390/diabetology4020015
    [Google Scholar]
  12. Avgerinos K.I. Spyrou N. Mantzoros C.S. Dalamaga M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism 2019 92 121 135 10.1016/j.metabol.2018.11.001 30445141
    [Google Scholar]
  13. Seiler A. Chen M.A. Brown R.L. Fagundes C.P. Obesity, dietary factors, nutrition, and breast cancer risk. Curr. Breast Cancer Rep. 2018 10 1 14 27 10.1007/s12609‑018‑0264‑0 30662586
    [Google Scholar]
  14. Yao X. Tian Z. Dyslipidemia and colorectal cancer risk: A meta-analysis of prospective studies. Cancer Causes Control 2015 26 2 257 268 10.1007/s10552‑014‑0507‑y 25488827
    [Google Scholar]
  15. Li C. Yang L. Zhang D. Jiang W. Systematic review and meta-analysis suggest that dietary cholesterol intake increases risk of breast cancer. Nutr. Res. 2016 36 7 627 635 10.1016/j.nutres.2016.04.009 27333953
    [Google Scholar]
  16. Santos R.C. Domingues G. Matias I. Matos J. Fonseca I. de Almeida J.M. Dias S. LDL-cholesterol signaling induces breast cancer proliferation and invasion. Lipids Health Dis. 2014 13 1 16 10.1186/1476‑511X‑13‑16 24428917
    [Google Scholar]
  17. Antalis C.J. Uchida A. Buhman K.K. Siddiqui R.A. Migration of MDA-MB-231 breast cancer cells depends on the availability of exogenous lipids and cholesterol esterification. Clin. Exp. Metastasis 2011 28 8 733 741 10.1007/s10585‑011‑9405‑9 21744083
    [Google Scholar]
  18. Guan X. Liu Z. Zhao Z. Zhang X. Tao S. Yuan B. Zhang J. Wang D. Liu Q. Ding Y. Emerging roles of low-density lipoprotein in the development and treatment of breast cancer. Lipids Health Dis. 2019 18 1 137 10.1186/s12944‑019‑1075‑7 31182104
    [Google Scholar]
  19. Lu C.W. Lo Y.H. Chen C.H. Lin C.Y. Tsai C.H. Chen P.J. Yang Y.F. Wang C.H. Tan C.H. Hou M.F. Yuan S.S.F. VLDL and LDL, but not HDL, promote breast cancer cell proliferation, metastasis and angiogenesis. Cancer Lett. 2017 388 130 138 10.1016/j.canlet.2016.11.033 27940127
    [Google Scholar]
  20. Greife A. Tukova J. Steinhoff C. Scott S.D. Schulz W.A. Hatina J. Establishment and characterization of a bladder cancer cell line with enhanced doxorubicin resistance by mevalonate pathway activation. Tumour Biol. 2015 36 5 3293 3300 10.1007/s13277‑014‑2959‑9 25566959
    [Google Scholar]
  21. Hultsch S. Kankainen M. Paavolainen L. Kovanen R.M. Ikonen E. Kangaspeska S. Pietiäinen V. Kallioniemi O. Association of tamoxifen resistance and lipid reprogramming in breast cancer. BMC Cancer 2018 18 1 850 10.1186/s12885‑018‑4757‑z 30143015
    [Google Scholar]
  22. Weber P. Wagner M. Schneckenburger H. Cholesterol dependent uptake and interaction of doxorubicin in mcf-7 breast cancer cells. Int. J. Mol. Sci. 2013 14 4 8358 8366 10.3390/ijms14048358 23591847
    [Google Scholar]
  23. Guo L. Breast cancer heterogeneity and its implication in personalized precision therapy. Exp Hematol Oncol 2023 12 1 3 10.1186/s40164‑022‑00363‑1
    [Google Scholar]
  24. Rakha E.A. Tse G.M. Quinn C.M. An update on the pathological classification of breast cancer. Histopathology 2023 82 1 5 16 10.1111/his.14786 36482272
    [Google Scholar]
  25. Burstein H.J. Temin S. Anderson H. Buchholz T.A. Davidson N.E. Gelmon K.E. Giordano S.H. Hudis C.A. Rowden D. Solky A.J. Stearns V. Winer E.P. Griggs J.J. Adjuvant endocrine therapy for women with hormone receptor-positive breast cancer: American society of clinical oncology clinical practice guideline focused update. J. Clin. Oncol. 2014 32 21 2255 2269 10.1200/JCO.2013.54.2258 24868023
    [Google Scholar]
  26. Koboldt D.C. Fulton R.S. McLellan M.D. Schmidt H. Veizer K.J. McMichael J.F. Fulton L.L. Dooling D.J. Ding L. Mardis E.R. Comprehensive molecular portraits of human breast tumours. Nature 2012 490 7418 61 70 10.1038/nature11412 23000897
    [Google Scholar]
  27. Yersal O. Barutca S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014 5 3 412 424 10.5306/wjco.v5.i3.412 25114856
    [Google Scholar]
  28. Yoon J. Oh D.Y. HER2-targeted therapies beyond breast cancer — An update. Nat. Rev. Clin. Oncol. 2024 21 9 675 700 10.1038/s41571‑024‑00924‑9 39039196
    [Google Scholar]
  29. Baranova A. Triple-negative breast cancer: Current treatment strategies and factors of negative prognosis. J Med Life. 2022 15 2 153 161 10.25122/jml‑2021‑0108
    [Google Scholar]
  30. Haukaas T.H. Euceda L.R. Giskeødegård G.F. Lamichhane S. Krohn M. Jernström S. Aure M.R. Lingjærde O.C. Schlichting E. Garred Ø. Due E.U. Mills G.B. Sahlberg K.K. Dale B.A.L. Bathen T.F. Metabolic clusters of breast cancer in relation to gene- and protein expression subtypes. Cancer Metab. 2016 4 1 12 10.1186/s40170‑016‑0152‑x 27350877
    [Google Scholar]
  31. Cappelletti V. Metabolic footprints and molecular subtypes in breast cancer. Dis Markers. 2017 2017 7687851
    [Google Scholar]
  32. Kim S. Differential expression of lipid metabolism-related proteins in different breast cancer subtypes. PloS one 2015 2015 e0119473
    [Google Scholar]
  33. Monaco M.E. Fatty acid metabolism in breast cancer subtypes. Oncotarget. 2017 8 17 29487 29500 10.18632/oncotarget.15494
    [Google Scholar]
  34. Luo J. Yang H. Song B.L. Mechanisms and regulation of cholesterol homeostasis. Nat. Rev. Mol. Cell Biol. 2020 21 4 225 245 10.1038/s41580‑019‑0190‑7 31848472
    [Google Scholar]
  35. Wang Y. Yutuc E. Griffiths W.J. Cholesterol metabolism pathways – Are the intermediates more important than the products? FEBS J. 2021 288 12 3727 3745 10.1111/febs.15727 33506652
    [Google Scholar]
  36. Goldstein J.L. Brown M.S. The ldl receptor and the regulation of cellular cholesterol metabolism. J. Cell Sci. 1985 1985 Suppl. 3 131 137 10.1242/jcs.1985.Supplement_3.13 3914990
    [Google Scholar]
  37. Maiti B. Kundranda M.N. Spiro T.P. Daw H.A. The association of metabolic syndrome with triple-negative breast cancer. Breast Cancer Res. Treat. 2010 121 2 479 483 10.1007/s10549‑009‑0591‑y 19851862
    [Google Scholar]
  38. DuSell C.D. Umetani M. Shaul P.W. Mangelsdorf D.J. McDonnell D.P. 27-hydroxycholesterol is an endogenous selective estrogen receptor modulator. Mol. Endocrinol. 2008 22 1 65 77 10.1210/me.2007‑0383 17872378
    [Google Scholar]
  39. Simigdala N. Gao Q. Pancholi S. Larsen R.H. Zvelebil M. Ribas R. Folkerd E. Thompson A. Bhamra A. Dowsett M. Martin L.A. Cholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor-positive breast cancer. Breast Cancer Res. 2016 18 1 58 10.1186/s13058‑016‑0713‑5 27246191
    [Google Scholar]
  40. Győrffy B. Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer. Comput. Struct. Biotechnol. J. 2021 19 4101 4109 10.1016/j.csbj.2021.07.014 34527184
    [Google Scholar]
  41. Tomczak K. Czerwińska P. Wiznerowicz M. Review the cancer genome atlas (TCGA): An immeasurable source of knowledge. Contemp Oncol 2015 19 1A A68 77 10.5114/wo.2014.47136
    [Google Scholar]
  42. Lánczky A. Győrffy B. Web-based survival analysis tool tailored for medical research (KMplot): Development and implementation. J. Med. Internet Res. 2021 23 7 e27633 10.2196/27633 34309564
    [Google Scholar]
  43. Benjamini Y. Hochberg Y. Controlling the false discovery rate: A practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 1995 57 1 289 300 10.1111/j.2517‑6161.1995.tb02031.x
    [Google Scholar]
  44. Love M.I. Huber W. Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014 15 12 550 10.1186/s13059‑014‑0550‑8 25516281
    [Google Scholar]
  45. Kim H.Y. Statistical notes for clinical researchers: Chi-squared test and Fisher’s exact test. Restor. Dent. Endod 2017 42 2 152 155 10.5395/rde.2017.42.2.152 28503482
    [Google Scholar]
  46. Seabold S. Perktold J. Statsmodels: Econometric and statistical modeling with python. SciPy 2010 7 92 96 10.25080/Majora‑92bf1922‑011
    [Google Scholar]
  47. Abdi H. Williams L.J. Tukey’s honestly significant difference (HSD) test. Encycl. Res. Des. 2010 0 1566 1570
    [Google Scholar]
  48. Wu S.Z. Al-Eryani G. Roden D.L. Junankar S. Harvey K. Andersson A. Thennavan A. Wang C. Torpy J.R. Bartonicek N. Wang T. Larsson L. Kaczorowski D. Weisenfeld N.I. Uytingco C.R. Chew J.G. Bent Z.W. Chan C.L. Gnanasambandapillai V. Dutertre C.A. Gluch L. Hui M.N. Beith J. Parker A. Robbins E. Segara D. Cooper C. Mak C. Chan B. Warrier S. Ginhoux F. Millar E. Powell J.E. Williams S.R. Liu X.S. O’Toole S. Lim E. Lundeberg J. Perou C.M. Swarbrick A. A single-cell and spatially resolved atlas of human breast cancers. Nat. Genet. 2021 53 9 1334 1347 10.1038/s41588‑021‑00911‑1 34493872
    [Google Scholar]
  49. Hao Y. Stuart T. Kowalski M.H. Choudhary S. Hoffman P. Hartman A. Srivastava A. Molla G. Madad S. Granda F.C. Satija R. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat. Biotechnol. 2023 42 2 293 304 10.1038/s41587‑023‑01767‑y 37231261
    [Google Scholar]
  50. Walsh C.A. The mevalonate precursor enzyme HMGCS1 is a novel marker and key mediator of cancer stem cell enrichment in luminal and basal models of breast cancer. PLoS One 2020 15 7 e0236187 10.1371/journal.pone.0236187
    [Google Scholar]
  51. Göbel A. Cholesterol and beyond-The role of the mevalonate pathway in cancer biology. Biochim Biophys Acta Rev Cancer 2020 1873 2 188351
    [Google Scholar]
  52. Bloch K. The biological synthesis of cholesterol. Science 1965 150 3692 19 28 10.1126/science.150.3692.19 5319508
    [Google Scholar]
  53. Mitsche M.A. McDonald J.G. Hobbs H.H. Cohen J.C. Flux analysis of cholesterol biosynthesis in vivo reveals multiple tissue and cell-type specific pathways. eLife 2015 4 e07999 10.7554/eLife.07999 26114596
    [Google Scholar]
  54. Xian M. Wang Q. Xiao L. Zhong L. Xiong W. Ye L. Su P. Zhang C. Li Y. Orlowski R.Z. Zhan F. Ganguly S. Zu Y. Qian J. Yi Q. Leukocyte immunoglobulin-like receptor B1 (LILRB1) protects human multiple myeloma cells from ferroptosis by maintaining cholesterol homeostasis. Nat. Commun. 2024 15 1 5767 10.1038/s41467‑024‑50073‑x 38982045
    [Google Scholar]
  55. Wu Y.X. Wang Y. Progress on the molecular mechanisms of PCSK9-mediated degradation of low density lipoprotein receptor. Yi Chuan 2020 42 10 965 978
    [Google Scholar]
  56. Sharpe L.J. Brown A.J. Controlling cholesterol synthesis beyond 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR). J. Biol. Chem. 2013 288 26 18707 18715 10.1074/jbc.R113.479808 23696639
    [Google Scholar]
  57. Borgquist S. Djerbi S. Pontén F. Anagnostaki L. Goldman M. Gaber A. Manjer J. Landberg G. Jirström K. HMG-CoA reductase expression in breast cancer is associated with a less aggressive phenotype and influenced by anthropometric factors. Int. J. Cancer 2008 123 5 1146 1153 10.1002/ijc.23597 18528862
    [Google Scholar]
  58. Lüönd F. Tiede S. Christofori G. Breast cancer as an example of tumour heterogeneity and tumour cell plasticity during malignant progression. Br. J. Cancer 2021 125 2 164 175 10.1038/s41416‑021‑01328‑7 33824479
    [Google Scholar]
  59. Ye F. Cancer-associated fibroblasts facilitate breast cancer progression through exosomal circTBPL1-mediated intercellular communication. Cell Death Dis 2023 14 7 471 10.1038/s41419‑023‑05986‑8
    [Google Scholar]
  60. Gao Y. CD63+ cancer-associated fibroblasts confer tamoxifen resistance to breast cancer cells through exosomal miR-22. Adv Sci 2020 7 21 2002518 10.1002/advs.202002518
    [Google Scholar]
  61. Rivas E.I. Targeted immunotherapy against distinct cancer-associated fibroblasts overcomes treatment resistance in refractory HER2+ breast tumors. Nat Commun 2022 13 1 5310 10.1038/s41467‑022‑32782‑3
    [Google Scholar]
  62. Santos Z.D. de Souza J.C. Pimenta T.M. Martins S.B. Junior R.S.R. Butzene S.M.S. Tessarolo N.G. Cilas P.M.L. Jr Silva I.V. Rangel L.B.A. The impact of lipid metabolism on breast cancer: A review about its role in tumorigenesis and immune escape. Cell Commun. Signal. 2023 21 1 161 10.1186/s12964‑023‑01178‑1 37370164
    [Google Scholar]
  63. Ehmsen S. Pedersen M.H. Wang G. Terp M.G. Arslanagic A. Hood B.L. Conrads T.P. Larsen L.R. Ditzel H.J. Increased cholesterol biosynthesis is a key characteristic of breast cancer stem cells influencing patient outcome. Cell Rep. 2019 27 13 3927 3938.e6 10.1016/j.celrep.2019.05.104 31242424
    [Google Scholar]
  64. O’Grady S. Crown J. Duffy M.J. Statins inhibit proliferation and induce apoptosis in triple-negative breast cancer cells. Med Oncol 2022 39 10 142 10.1007/s12032‑022‑01733‑9
    [Google Scholar]
  65. Santos Z.D. Guimaraes S.I. Sanni H.M.F. Cochran B.J. Rye K.A. Grewal T. Hoy A.J. Rangel L.B.A. Atorvastatin improves cisplatin sensitivity through modulation of cholesteryl ester homeostasis in breast cancer cells. Discov. Oncol. 2022 13 1 135 10.1007/s12672‑022‑00598‑8 36481936
    [Google Scholar]
  66. Sim Y. Lim C. Phyu N. Tan K.T.B. Chew L.S.T. Wong C.Y. Madhukumar P. Yong W.S. Lim S.Z. Hamzah J.L.B. Tan S.Y. Chay W.Y. Wong F.Y. Tan P.H. Tan V.K.M. The impact of statin use and breast cancer recurrence-A retrospective study in Singapore. Front. Oncol. 2022 12 835320 10.3389/fonc.2022.835320 35433431
    [Google Scholar]
  67. Sinha K.C. Ignatoski K.W. Lippman M.E. Ethier S.P. Chinnaiyan A.M. Transcriptome analysis of HER2 reveals a molecular connection to fatty acid synthesis. Cancer Res. 2003 63 1 132 139 12517789
    [Google Scholar]
  68. Calvo G.D. Vilaró L.L. Nasarre L. Olabarria P.M. Vázquez T. Escuin D. Badimon L. Barnadas A. Lerma E. Cortés L.V. Intratumor cholesteryl ester accumulation is associated with human breast cancer proliferation and aggressive potential: A molecular and clinicopathological study. BMC Cancer 2015 15 1 460 10.1186/s12885‑015‑1469‑5 26055977
    [Google Scholar]
  69. Martin H.G. Rivas L.A. Statins activate a mitochondria-operated pathway of apoptosis in breast tumor cells by a mechanism regulated by ErbB2 and dependent on the prenylation of proteins. FEBS Lett. 2008 582 17 2589 2594 10.1016/j.febslet.2008.06.034 18582466
    [Google Scholar]
  70. Zhang J. Li Q. Wu Y. Wang D. Xu L. Zhang Y. Wang S. Wang T. Liu F. Zaky M.Y. Hou S. Liu S. Zou K. Lei H. Zou L. Zhang Y. Liu H. Cholesterol content in cell membrane maintains surface levels of ErbB2 and confers a therapeutic vulnerability in ErbB2-positive breast cancer. Cell Commun. Signal. 2019 17 1 15 10.1186/s12964‑019‑0328‑4 30786890
    [Google Scholar]
  71. Li X. Tang H. Wang J. Xie X. Liu P. Kong Y. Ye F. Shuang Z. Xie Z. Xie X. The effect of preoperative serum triglycerides and high-density lipoprotein-cholesterol levels on the prognosis of breast cancer. Breast 2017 32 1 6 10.1016/j.breast.2016.11.024 27939967
    [Google Scholar]
  72. Santos R.C. Fonseca I. Dias S. Almeida M.J.C. Plasma level of LDL-cholesterol at diagnosis is a predictor factor of breast tumor progression. BMC Cancer 2014 14 1 132 10.1186/1471‑2407‑14‑132 24571647
    [Google Scholar]
  73. Wyhe V.R. Rahal O. Woodward W. Effect of statins on breast cancer recurrence and mortality: A review. Breast Cancer 2017 9 559 565 10.2147/BCTT.S148080 29238220
    [Google Scholar]
  74. Antalis C.J. Arnold T. Rasool T. Lee B. Buhman K.K. Siddiqui R.A. High ACAT1 expression in estrogen receptor negative basal-like breast cancer cells is associated with LDL-induced proliferation. Breast Cancer Res. Treat. 2010 122 3 661 670 10.1007/s10549‑009‑0594‑8 19851860
    [Google Scholar]
  75. Yuan Q. Lu X. Guo H. Sun J. Yang M. Liu Q. Tong M. Low-density lipoprotein receptor promotes crosstalk between cell stemness and tumor immune microenvironment in breast cancer: A large data-based multi-omics study. J. Transl. Med. 2023 21 1 871 10.1186/s12967‑023‑04699‑y 38037058
    [Google Scholar]
  76. Hsu C.Y. Abdulrahim M.N. Mustafa M.A. Omar T.M. Balto F. Pineda I. Khudair T.T. Ubaid M. Ali M.S. The multifaceted role of PCSK9 in cancer pathogenesis, tumor immunity, and immunotherapy. Med. Oncol. 2024 41 8 202 10.1007/s12032‑024‑02435‑0 39008137
    [Google Scholar]
  77. Chong W.E. Joncas F.H. Seidah N.G. Calon F. Diorio C. Gangloff A. Circulating levels of PCSK9, ANGPTL3 and Lp(A) in stage III breast cancers. BMC Cancer 2022 22 1 1049 10.1186/s12885‑022‑10120‑6 36203122
    [Google Scholar]
  78. Llaverias G. Danilo C. Mercier I. Daumer K. Capozza F. Williams T.M. Sotgia F. Lisanti M.P. Frank P.G. Role of cholesterol in the development and progression of breast cancer. Am. J. Pathol. 2011 178 1 402 412 10.1016/j.ajpath.2010.11.005 21224077
    [Google Scholar]
  79. Nowak C. Ärnlöv J. A Mendelian randomization study of the effects of blood lipids on breast cancer risk. Nat Commun 2018 9 1 3957 10.1038/s41467‑018‑06467‑9
    [Google Scholar]
  80. Borojeni M.A.A. Nik E.M. Jaafari R.M. Banach M. Sahebkar A. Effects of immunization against PCSK9 in an experimental model of breast cancer. Arch. Med. Sci. 2019 15 3 570 579 10.5114/aoms.2019.84734 31110521
    [Google Scholar]
  81. Sun Y. Zhang H. Meng J. Guo F. Ren D. Wu H. Jin X. S-palmitoylation of PCSK9 induces sorafenib resistance in liver cancer by activating the PI3K/AKT pathway. Cell Rep. 2022 40 7 111194 10.1016/j.celrep.2022.111194 35977495
    [Google Scholar]
  82. Yang K. Zhu J. Luo H. Yu S. Wang L. Pro-protein convertase subtilisin/kexin type 9 promotes intestinal tumor development by activating Janus kinase 2/signal transducer and activator of transcription 3/SOCS3 signaling in Apc Min/+ mice. Int. J. Immunopathol. Pharmacol. 2021 35 20587384211038345 10.1177/20587384211038345 34586888
    [Google Scholar]
  83. Jin W. Role of JAK/STAT3 signaling in the regulation of metastasis, the transition of cancer stem cells, and chemoresistance of cancer by epithelial–mesenchymal transition. Cells 2020 9 1 217 10.3390/cells9010217
    [Google Scholar]
  84. Bernardi S. Marcuzzi A. Piscianz E. Tommasini A. Fabris B. The complex interplay between lipids, immune system and interleukins in cardio-metabolic diseases. Int. J. Mol. Sci. 2018 19 12 4058 10.3390/ijms19124058 30558209
    [Google Scholar]
  85. Liu X. Bao X. Hu M. Chang H. Jiao M. Cheng J. Xie L. Huang Q. Li F. Li C.Y. Inhibition of PCSK9 potentiates immune checkpoint therapy for cancer. Nature 2020 588 7839 693 698 10.1038/s41586‑020‑2911‑7 33177715
    [Google Scholar]
  86. Yuan J. Cai T. Zheng X. Ren Y. Qi J. Lu X. Chen H. Lin H. Chen Z. Liu M. He S. Chen Q. Feng S. Wu Y. Zhang Z. Ding Y. Yang W. Potentiating CD8+ T cell antitumor activity by inhibiting PCSK9 to promote LDLR-mediated TCR recycling and signaling. Protein Cell 2021 12 4 240 260 10.1007/s13238‑021‑00821‑2 33606190
    [Google Scholar]
  87. Lei K. Kurum A. Kaynak M. Bonati L. Han Y. Cencen V. Gao M. Xie Y.Q. Guo Y. Hannebelle M.T.M. Wu Y. Zhou G. Guo M. Fantner G.E. Sakar M.S. Tang L. Cancer-cell stiffening via cholesterol depletion enhances adoptive T-cell immunotherapy. Nat. Biomed. Eng. 2021 5 12 1411 1425 10.1038/s41551‑021‑00826‑6 34873307
    [Google Scholar]
  88. Wang R. Liu H. He P. An D. Guo X. Zhang X. Feng M. Inhibition of PCSK9 enhances the antitumor effect of PD-1 inhibitor in colorectal cancer by promoting the infiltration of CD8+ T cells and the exclusion of Treg cells. Front. Immunol. 2022 13 947756 10.3389/fimmu.2022.947756 36003387
    [Google Scholar]
  89. Wang Lu PCSK9 promotes the progression and metastasis of colon cancer cells through regulation of EMT and PI3K/AKT signaling in tumor cells and phenotypic polarization of macrophages. J Exp Clin Cancer Res 2022 41 1 303 10.1186/s13046‑022‑02477‑0
    [Google Scholar]
  90. Lebeau P.F. Platko K. Byun J.H. Makda Y. Austin R.C. The emerging roles of intracellular PCSK9 and their implications in endoplasmic reticulum stress and metabolic diseases. Metabolites 2022 12 3 215 10.3390/metabo12030215 35323658
    [Google Scholar]
/content/journals/cmc/10.2174/0109298673291217241219055416
Loading
/content/journals/cmc/10.2174/0109298673291217241219055416
Loading

Data & Media loading...

Supplements

Supplementary material is available on the publisher's website along with the published article.


  • Article Type:
    Research Article
Keywords: triple-negative ; PCSK9 ; Breast cancer ; cholesterol ; HER2+ ; LDLR ; basal-like
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test