The feasibility of using an autologous GM-CSF-secreting breast cancer vaccine to induce immunity in patients with stage II-III and metastatic breast cancers

Karen S Anderson, Timothy K Erick, Meixuan Chen, Heather Daley, Margaret Campbell, Yolonda Colson, Martin Mihm, Labib R Zakka, Marika Hopper, William Barry, Eric P Winer, Glenn Dranoff, Beth Overmoyer, Karen S Anderson, Timothy K Erick, Meixuan Chen, Heather Daley, Margaret Campbell, Yolonda Colson, Martin Mihm, Labib R Zakka, Marika Hopper, William Barry, Eric P Winer, Glenn Dranoff, Beth Overmoyer

Abstract

Purpose: The antigenic targets of immunity and the role of vaccination in breast cancer are unknown. We performed a phase I study of an autologous GM-CSF-secreting breast cancer vaccine in patients with metastatic and stage II-III breast cancer.

Methods: Tumor cells from patients with metastatic (n = 15) and stage II-III (n = 7) disease were transduced with a replication-defective adenoviral vector encoding GM-CSF, and then irradiated. Twelve and seven patients with metastatic and stage II-III disease, respectively, received weekly vaccination for three weeks, followed by every other week until disease progression or vaccine supply was exhausted (metastatic) or until six total vaccine doses were administered (stage II-III).

Results: Among those patients with metastatic disease who received vaccinations, eight had progressive disease at two months, three had stable disease for 4-13 months, and one has had no evidence of disease for 13 years. Of the patients with stage II-III disease, five died of metastatic disease between 1.16 and 8.49 years after the start of vaccinations (median 6.24 years) and two are alive as of September 2021. Toxicities included injection site reactions, fatigue, fever, upper respiratory symptoms, joint pain, nausea, and edema. Four of five evaluable patients with metastatic disease developed a skin reaction with immune cell infiltration after the fifth injection of unmodified, irradiated tumor cells.

Conclusion: We conclude that tumor cells can be harvested from patients with metastatic or stage II-III breast cancer to prepare autologous GM-CSF-secreting vaccines that induce coordinated immune responses with limited toxicity. TRIAL REGISTRATION AND DATE OF REGISTRATION: clinicaltrials.gov, NCT00317603 (April 25, 2006) and NCT00880464 (April 13, 2009).

Keywords: Autologous cellular vaccine; Breast cancer; GM-CSF.

Conflict of interest statement

GD is an employee of Novartis with stock ownership.

© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.

Figures

Fig. 1
Fig. 1
Vaccination induces local inflammation in patients with metastatic breast cancer. a GM-CSF-modified tumor cells were injected in contralateral limbs in patients with metastatic breast cancer. Skin site reactions were observed in seven patients 48–72 h after the first vaccine dose and in five patients after the fifth vaccine dose, and the longest dimension of erythema (cm) is shown, with patient numbers noted. Other patients did not exhibit any skin reaction. b Average baseline erythema at each vaccine dose level. c Unmodified cells at 106 cells/dose (DTH) were injected in contralateral limbs of seven patients with metastatic breast cancer. Skin site reactions were measured 48–72 h after the first and fifth injection. No responses were observed after the first injection. Four responses were observed after the fifth injection, and the longest dimension of erythema (cm) is shown, with patient numbers noted
Fig. 2
Fig. 2
Vaccination induces leukocyte infiltration. Representative skin biopsies from a patient with metastatic breast cancer showing inflammatory cellular infiltration following a injection of GVAX vaccine, and b following the fifth dose of 1 × 106 non-transduced, irradiated cells (DTH). Inflammatory infiltrate included lymphocytes, granulocytes, and macrophages
Fig. 3
Fig. 3
Immune profile for selected patients with metastatic breast cancer who received GVAX vaccine. a Immune profile among patients with stable disease or disease response following vaccination. b Immune profile among patients with disease progression following vaccination. Heatmaps of antibody response (normalized signal > 1.5) in vaccinated patients compared with healthy controls. D = Day number post-first GVAX vaccination

References

    1. Ward JP, Gubin MM, Schreiber RD. The role of neoantigens in naturally occurring and therapeutically induced immune responses to cancer. Adv Immunol. 2016;130:25–74. doi: 10.1016/bs.ai.2016.01.001.
    1. Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccin. 2019;4:7. doi: 10.1038/s41541-019-0103-y.
    1. Sahin U, Tureci O. Personalized vaccines for cancer immunotherapy. Science. 2018;359(6382):1355–1360. doi: 10.1126/science.aar7112.
    1. Kaufman HL, Ruby CE, Hughes T, Slingluff CL., Jr Current status of granulocyte-macrophage colony-stimulating factor in the immunotherapy of melanoma. J Immunother Cancer. 2014;2:11. doi: 10.1186/2051-1426-2-11.
    1. Dranoff G, Jaffee E, Lazenby A, Golumbek P, Levitsky H, Brose K, Jackson V, Hamada H, Pardoll D, Mulligan RC. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci USA. 1993;90(8):3539–3543. doi: 10.1073/pnas.90.8.3539.
    1. Small EJ, Sacks N, Nemunaitis J, Urba WJ, Dula E, Centeno AS, Nelson WG, Ando D, Howard C, Borellini F, Nguyen M, Hege K, Simons JW. Granulocyte macrophage colony-stimulating factor–secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clin Cancer Res. 2007;13(13):3883–3891. doi: 10.1158/1078-0432.CCR-06-2937.
    1. Laheru D, Yeo C, Biedrzycki B, Solt S, Lutz E, Onners B, Tartakovsky I, Herman J, Hruban R, Piantadosi S, Jaffee E. A safety and efficacy trial of lethally irradiated allogeneic pancreatic tumor cells transfected with the GM-CSF gene in combination with adjuvant chemoradiotherapy for the treatment of adenocarcinoma of the pancreas. J Clin Oncol. 2007;25(18):3010–3010. doi: 10.1200/jco.2007.25.18_suppl.3010.
    1. Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, Stearns V, Disis ML, Ye X, Piantadosi S, Fetting JH, Davidson NE, Jaffee EM. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol. 2009;27(35):5911–5918. doi: 10.1200/JCO.2009.23.3494.
    1. Chen G, Gupta R, Petrik S, Laiko M, Leatherman JM, Asquith JM, Daphtary MM, Garrett-Mayer E, Davidson NE, Hirt K, Berg M, Uram JN, Dauses T, Fetting J, Duus EM, Atay-Rosenthal S, Ye X, Wolff AC, Stearns V, Jaffee EM, Emens LA. A feasibility study of cyclophosphamide, trastuzumab, and an allogeneic GM-CSF-secreting breast tumor vaccine for HER2+ metastatic breast cancer. Cancer Immunol Res. 2014;2(10):949–961. doi: 10.1158/2326-6066.CIR-14-0058.
    1. Salgia R, Lynch T, Skarin A, Lucca J, Lynch C, Jung K, Hodi FS, Jaklitsch M, Mentzer S, Swanson S, Lukanich J, Bueno R, Wain J, Mathisen D, Wright C, Fidias P, Donahue D, Clift S, Hardy S, Neuberg D, Mulligan R, Webb I, Sugarbaker D, Mihm M, Dranoff G. Vaccination with irradiated autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor augments antitumor immunity in some patients with metastatic non-small-cell lung carcinoma. J Clin Oncol. 2003;21(4):624–630. doi: 10.1200/JCO.2003.03.091.
    1. Soiffer R, Lynch T, Mihm M, Jung K, Rhuda C, Schmollinger JC, Hodi FS, Liebster L, Lam P, Mentzer S, Singer S, Tanabe KK, Cosimi AB, Duda R, Sober A, Bhan A, Daley J, Neuberg D, Parry G, Rokovich J, Richards L, Drayer J, Berns A, Clift S, Cohen LK, Mulligan RC, Dranoff G. Vaccination with irradiated autologous melanoma cells engineered to secrete human granulocyte-macrophage colony-stimulating factor generates potent antitumor immunity in patients with metastatic melanoma. Proc Natl Acad Sci USA. 1998;95(22):13141–13146. doi: 10.1073/pnas.95.22.13141.
    1. Nemunaitis J, Murray N. Immune-modulating vaccines in non-small cell lung cancer. J Thorac Oncol. 2006;1(7):756–761.
    1. Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, Nikolskaya T, Serebryiskaya T, Beroukhim R, Hu M, Halushka MK, Sukumar S, Parker LM, Anderson KS, Harris LN, Garber JE, Richardson AL, Schnitt SJ, Nikolsky Y, Gelman RS, Polyak K. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007;11(3):259–273. doi: 10.1016/j.ccr.2007.01.013.
    1. Soiffer R, Hodi FS, Haluska F, Jung K, Gillessen S, Singer S, Tanabe K, Duda R, Mentzer S, Jaklitsch M, Bueno R, Clift S, Hardy S, Neuberg D, Mulligan R, Webb I, Mihm M, Dranoff G. Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma. J Clin Oncol. 2003;21(17):3343–3350. doi: 10.1200/JCO.2003.07.005.
    1. Anderson KS, Ramachandran N, Wong J, Raphael JV, Hainsworth E, Demirkan G, Cramer D, Aronzon D, Hodi FS, Harris L, Logvinenko T, LaBaer J. Application of protein microarrays for multiplexed detection of antibodies to tumor antigens in breast cancer. J Proteome Res. 2008;7(4):1490–1499. doi: 10.1021/pr700804c.
    1. Song L, Wiktor P, Qiu J, LaBaer J. Identification of antibody biomarker using high-density nucleic acid programmable protein array. Methods Mol Biol. 2021;2344:47–64. doi: 10.1007/978-1-0716-1562-1_4.
    1. Witt AE, Hines LM, Collins NL, Hu Y, Gunawardane RN, Moreira D, Raphael J, Jepson D, Koundinya M, Rolfs A, Taron B, Isakoff SJ, Brugge JS, LaBaer J. Functional proteomics approach to investigate the biological activities of cDNAs implicated in breast cancer. J Proteome Res. 2006;5(3):599–610. doi: 10.1021/pr050395r.
    1. Katchman BA, Chowell D, Wallstrom G, Vitonis AF, LaBaer J, Cramer DW, Anderson KS. Autoantibody biomarkers for the detection of serous ovarian cancer. Gynecol Oncol. 2017;146(1):129–136. doi: 10.1016/j.ygyno.2017.04.005.
    1. Curry WT, Jr, Gorrepati R, Piesche M, Sasada T, Agarwalla P, Jones PS, Gerstner ER, Golby AJ, Batchelor TT, Wen PY, Mihm MC, Dranoff G. Vaccination with irradiated autologous tumor cells mixed with irradiated GM-K562 cells stimulates antitumor immunity and T lymphocyte activation in patients with recurrent malignant glioma. Clin Cancer Res. 2016;22(12):2885–2896. doi: 10.1158/1078-0432.CCR-15-2163.
    1. Hodi FS, Butler M, Oble DA, Seiden MV, Haluska FG, Kruse A, Macrae S, Nelson M, Canning C, Lowy I, Korman A, Lautz D, Russell S, Jaklitsch MT, Ramaiya N, Chen TC, Neuberg D, Allison JP, Mihm MC, Dranoff G. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci USA. 2008;105(8):3005–3010. doi: 10.1073/pnas.0712237105.
    1. Goldberg JM, Fisher DE, Demetri GD, Neuberg D, Allsop SA, Fonseca C, Nakazaki Y, Nemer D, Raut CP, George S, Morgan JA, Wagner AJ, Freeman GJ, Ritz J, Lezcano C, Mihm M, Canning C, Hodi FS, Dranoff G. Biologic activity of autologous, granulocyte-macrophage colony-stimulating factor secreting alveolar soft-part sarcoma and clear cell sarcoma vaccines. Clin Cancer Res. 2015;21(14):3178–3186. doi: 10.1158/1078-0432.CCR-14-2932.
    1. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, Davis T, Henry-Spires R, MacRae S, Willman A, Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman A, Allison JP, Dranoff G. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A. 2003;100(8):4712–4717. doi: 10.1073/pnas.0830997100.
    1. Dranoff G. GM-CSF-based cancer vaccines. Immunol Rev. 2002;188:147–154. doi: 10.1034/j.1600-065x.2002.18813.x.
    1. Kaaks R, Fortner RT, Husing A, Barrdahl M, Hopper M, Johnson T, Tjonneland A, Hansen L, Overvad K, Fournier A, Boutron-Ruault MC, Kvaskoff M, Dossus L, Johansson M, Boeing H, Trichopoulou A, Benetou V, La Vecchia C, Sieri S, Mattiello A, Palli D, Tumino R, Matullo G, Onland-Moret NC, Gram IT, Weiderpass E, Sanchez MJ, Navarro Sanchez C, Duell EJ, Ardanaz E, Larranaga N, Lundin E, Idahl A, Jirstrom K, Nodin B, Travis RC, Riboli E, Merritt M, Aune D, Terry K, Cramer DW, Anderson KS. Tumor-associated autoantibodies as early detection markers for ovarian cancer? A prospective evaluation. Int J Cancer. 2018;143(3):515–526. doi: 10.1002/ijc.31335.
    1. Hattori N, Nakayama Y, Kitagawa K, Ishihara T, Saiki Y, Inagaki C. Anti-prolactin (PRL) autoantibody-binding sites (epitopes) on PRL molecule in macroprolactinemia. J Endocrinol. 2006;190(2):287–293. doi: 10.1677/joe.1.06871.
    1. Weihrauch MR, Ansen S, Jurkiewicz E, Geisen C, Xia Z, Anderson KS, Gracien E, Schmidt M, Wittig B, Diehl V, Wolf J, Bohlen H, Nadler LM. Phase I/II combined chemoimmunotherapy with carcinoembryonic antigen-derived HLA-A2-restricted CAP-1 peptide and irinotecan, 5-fluorouracil, and leucovorin in patients with primary metastatic colorectal cancer. Clin Cancer Res. 2005;11(16):5993–6001. doi: 10.1158/1078-0432.CCR-05-0018.
    1. Maecker B, Sherr DH, Vonderheide RH, von Bergwelt-Baildon MS, Hirano N, Anderson KS, Xia Z, Butler MO, Wucherpfennig KW, O'Hara C, Cole G, Kwak SS, Ramstedt U, Tomlinson AJ, Chicz RM, Nadler LM, Schultze JL. The shared tumor-associated antigen cytochrome P450 1B1 is recognized by specific cytotoxic T cells. Blood. 2003;102(9):3287–3294. doi: 10.1182/blood-2003-05-1374.
    1. Rongcun Y, Salazar-Onfray F, Charo J, Malmberg KJ, Evrin K, Maes H, Kono K, Hising C, Petersson M, Larsson O, Lan L, Appella E, Sette A, Celis E, Kiessling R. Identification of new HER2/neu-derived peptide epitopes that can elicit specific CTL against autologous and allogeneic carcinomas and melanomas. J Immunol. 1999;163(2):1037–1044.
    1. Mitchell MS, Lund TA, Sewell AK, Marincola FM, Paul E, Schroder K, Wilson DB, Kan-Mitchell J. The cytotoxic T cell response to peptide analogs of the HLA-A*0201-restricted MUC1 signal sequence epitope, M12. Cancer Immunol Immunother. 2007;56(3):287–301. doi: 10.1007/s00262-006-0191-1.
    1. Held G, Matsuo M, Epel M, Gnjatic S, Ritter G, Lee SY, Tai TY, Cohen CJ, Old LJ, Pfreundschuh M, Reiter Y, Hoogenboom HR, Renner C. Dissecting cytotoxic T cell responses towards the NY-ESO-1 protein by peptide/MHC-specific antibody fragments. Eur J Immunol. 2004;34(10):2919–2929. doi: 10.1002/eji.200425297.
    1. Andersen MH, Pedersen LO, Capeller B, Brocker EB, Becker JC, thor Straten P, Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients. Cancer Res. 2001;61(16):5964–5968.
    1. Zhang GL, Khan AM, Srinivasan KN, August JT, Brusic V. MULTIPRED: a computational system for prediction of promiscuous HLA binding peptides. Nucleic Acids Res (Web Server issue) 2005 doi: 10.1093/nar/gki452.
    1. Mirshahidi S, Kramer VG, Whitney JB, Essono S, Lee S, Dranoff G, Anderson KS, Ruprecht RM. Overlapping synthetic peptides encoding TPD52 as breast cancer vaccine in mice: prolonged survival. Vaccine. 2009;27(12):1825–1833. doi: 10.1016/j.vaccine.2009.01.089.
    1. Porter D, Lahti-Domenici J, Keshaviah A, Bae YK, Argani P, Marks J, Richardson A, Cooper A, Strausberg R, Riggins GJ, Schnitt S, Gabrielson E, Gelman R, Polyak K. Molecular markers in ductal carcinoma in situ of the breast. Mol Cancer Res. 2003;1(5):362–375.
    1. Herr W, Ranieri E, Gambotto A, Kierstead LS, Amoscato AA, Gesualdo L, Storkus WJ. Identification of naturally processed and HLA-presented Epstein-Barr virus peptides recognized by CD4(+) or CD8(+) T lymphocytes from human blood. Proc Natl Acad Sci USA. 1999;96(21):12033–12038. doi: 10.1073/pnas.96.21.12033.
    1. Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, Tureci O, Sahin U. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res. 2010;70(22):9031–9040. doi: 10.1158/0008-5472.CAN-10-0699.
    1. Ott PA, Hu-Lieskovan S, Chmielowski B, Govindan R, Naing A, Bhardwaj N, Margolin K, Awad MM, Hellmann MD, Lin JJ, Friedlander T, Bushway ME, Balogh KN, Sciuto TE, Kohler V, Turnbull SJ, Besada R, Curran RR, Trapp B, Scherer J, Poran A, Harjanto D, Barthelme D, Ting YS, Dong JZ, Ware Y, Huang Y, Huang Z, Wanamaker A, Cleary LD, Moles MA, Manson K, Greshock J, Khondker ZS, Fritsch E, Rooney MS, DeMario M, Gaynor RB, Srinivasan L. A phase Ib trial of personalized neoantigen therapy plus anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. Cell. 2020;183(2):347–362. doi: 10.1016/j.cell.2020.08.053.
    1. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Lower M, Bukur V, Tadmor AD, Luxemburger U, Schrors B, Omokoko T, Vormehr M, Albrecht C, Paruzynski A, Kuhn AN, Buck J, Heesch S, Schreeb KH, Muller F, Ortseifer I, Vogler I, Godehardt E, Attig S, Rae R, Breitkreuz A, Tolliver C, Suchan M, Martic G, Hohberger A, Sorn P, Diekmann J, Ciesla J, Waksmann O, Bruck AK, Witt M, Zillgen M, Rothermel A, Kasemann B, Langer D, Bolte S, Diken M, Kreiter S, Nemecek R, Gebhardt C, Grabbe S, Holler C, Utikal J, Huber C, Loquai C, Tureci O. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547(7662):222–226. doi: 10.1038/nature23003.
    1. Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, Parkhurst MR, Yossef R, Lowery FJ, Jafferji MS, Prickett TD, Goff SL, McGowan CT, Seitter S, Shindorf ML, Parikh A, Chatani PD, Robbins PF, Rosenberg SA. mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J Clin Invest. 2020;130(11):5976–5988. doi: 10.1172/JCI134915.
    1. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168(4):707–723. doi: 10.1016/j.cell.2017.01.017.
    1. Reddish M, MacLean GD, Koganty RR, Kan-Mitchell J, Jones V, Mitchell MS, Longenecker BM. Anti-MUC1 class I restricted CTLs in metastatic breast cancer patients immunized with a synthetic MUC1 peptide. Int J Cancer. 1998;76(6):817–823. doi: 10.1002/(sici)1097-0215(19980610)76:6<817::aid-ijc9>;2-0.
    1. Knutson KL, Schiffman K, Disis ML. Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients. J Clin Invest. 2001;107(4):477–484. doi: 10.1172/JCI11752.
    1. Nair SK, Heiser A, Boczkowski D, Majumdar A, Naoe M, Lebkowski JS, Vieweg J, Gilboa E. Induction of cytotoxic T cell responses and tumor immunity against unrelated tumors using telomerase reverse transcriptase RNA transfected dendritic cells. Nat Med. 2000;6(9):1011–1017. doi: 10.1038/79519.
    1. O'Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 2019;16(3):151–167. doi: 10.1038/s41571-018-0142-8.
    1. Turajlic S, Sottoriva A, Graham T, Swanton C. Resolving genetic heterogeneity in cancer. Nat Rev Genet. 2019;20(7):404–416. doi: 10.1038/s41576-019-0114-6.
    1. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541–550. doi: 10.1038/s41591-018-0014-x.
    1. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, Swain SM, Prowell T, Loibl S, Wickerham DL, Bogaerts J, Baselga J, Perou C, Blumenthal G, Blohmer J, Mamounas EP, Bergh J, Semiglazov V, Justice R, Eidtmann H, Paik S, Piccart M, Sridhara R, Fasching PA, Slaets L, Tang S, Gerber B, Geyer CE, Jr, Pazdur R, Ditsch N, Rastogi P, Eiermann W, von Minckwitz G. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–172. doi: 10.1016/S0140-6736(13)62422-8.
    1. Anderson KS, Sibani S, Wallstrom G, Qiu J, Mendoza EA, Raphael J, Hainsworth E, Montor WR, Wong J, Park JG, Lokko N, Logvinenko T, Ramachandran N, Godwin AK, Marks J, Engstrom P, Labaer J. Protein microarray signature of autoantibody biomarkers for the early detection of breast cancer. J Proteome Res. 2011;10(1):85–96. doi: 10.1021/pr100686b.
    1. Bast RC, Jr, Lu Z, Han CY, Lu KH, Anderson KS, Drescher CW, Skates SJ. Biomarkers and strategies for early detection of ovarian cancer. Cancer Epidemiol Biomarkers Prev. 2020;29(12):2504–2512. doi: 10.1158/1055-9965.EPI-20-1057.
    1. Rauf F, Anderson KS, LaBaer J. Autoantibodies in early detection of breast cancer. Cancer Epidemiol Biomarkers Prev. 2020;29(12):2475–2485. doi: 10.1158/1055-9965.EPI-20-0331.
    1. Wang J, Figueroa JD, Wallstrom G, Barker K, Park JG, Demirkan G, Lissowska J, Anderson KS, Qiu J, LaBaer J. Plasma autoantibodies associated with basal-like breast cancers. Cancer Epidemiol Biomark Prev. 2015;24(9):1332–1340. doi: 10.1158/1055-9965.EPI-15-0047.
    1. Schmid P, Cortes J, Pusztai L, McArthur H, Kummel S, Bergh J, Denkert C, Park YH, Hui R, Harbeck N, Takahashi M, Foukakis T, Fasching PA, Cardoso F, Untch M, Jia L, Karantza V, Zhao J, Aktan G, Dent R, O'Shaughnessy J, Investigators K. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382(9):810–821. doi: 10.1056/NEJMoa1910549.
    1. Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, Gallardo C, Lipatov O, Barrios CH, Holgado E, Iwata H, Masuda N, Otero MT, Gokmen E, Loi S, Guo Z, Zhao J, Aktan G, Karantza V, Schmid P. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396(10265):1817–1828. doi: 10.1016/s0140-6736(20)32531-9.

Source: PubMed

3
Subscribe