TH1902, a new docetaxel-peptide conjugate for the treatment of sortilin-positive triple-negative breast cancer

Michel Demeule, Cyndia Charfi, Jean-Christophe Currie, Alain Larocque, Alain Zgheib, Sophie Kozelko, Richard Béliveau, Christian Marsolais, Borhane Annabi, Michel Demeule, Cyndia Charfi, Jean-Christophe Currie, Alain Larocque, Alain Zgheib, Sophie Kozelko, Richard Béliveau, Christian Marsolais, Borhane Annabi

Abstract

Triple-negative breast cancer (TNBC) is a heterogeneous subgroup of cancers which lacks the expression and/or amplification of targetable biomarkers (ie, estrogen receptor, progestrogen receptor, and human epidermal growth factor receptor 2), and is often associated with the worse disease-specific outcomes than other breast cancer subtypes. Here, we report that high expression of the sortilin (SORT1) receptor correlates with the decreased survival in TNBC patients, and more importantly in those bearing lymph node metastases. By exploiting SORT1 function in ligand internalization, a new anticancer treatment strategy was designed to target SORT1-positive TNBC-derived cells both in vitro and in two in vivo tumor xenografts models. A peptide (TH19P01), which requires SORT1 for internalization and to which many anticancer drugs could be conjugated, was developed. In vitro, while the TH19P01 peptide itself did not exert any antiproliferative or apoptotic effects, the docetaxel-TH19P01 conjugate (TH1902) exerted potent antiproliferative and antimigratory activities when tested on TNBC-derived MDA-MB-231 cells. TH1902 triggered faster and more potent apoptotic cell death than did unconjugated docetaxel. The apoptotic and antimigratory effects of TH1902 were both reversed by two SORT1 ligands, neurotensin and progranulin, and on siRNA-mediated silencing of SORT1. TH1902 also altered microtubule polymerization and triggered the downregulation of the anti-apoptotic Bcl-xL biomarker. In vivo, both i.p. and i.v. administrations of TH1902 led to greater tumor regression in two MDA-MB-231 and HCC-70 murine xenograft models than did docetaxel, without inducing neutropenia. Altogether, the data demonstrates the high in vivo efficacy and safety of TH1902 against TNBC through a SORT1 receptor-mediated mechanism. This property allows for selective treatment of SORT1-positive TNBC and makes TH1902 a promising avenue for personalized therapy with the potential of improving the therapeutic window of cytotoxic anticancer drugs such as docetaxel.

Keywords: docetaxel; peptide-drug conjugate; receptor-mediated chemotherapy; sortilin; triple-negative breast cancer.

Conflict of interest statement

MD, AL, RB and BA were scientific founders of Katana Biopharma. CM is senior vice president and chief medical officer at Theratechnologies.

© 2021 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

FIGURE 1
FIGURE 1
SORT1 expression in breast cancers. A, Immunohistochemistry stainings were performed to assess SORT1 expression in normal adjacent tissue and in stage IIIC infiltrating duct carcinoma (IDC) and involved lymph node carcinoma (ILNC). B, SORT1 staining intensity was evaluated using the immunohistostaining (IHS) method on nine normal adjacent tissues, 35 IDC, 10 ILNC, and 19 TNBC tumors. Data are represented as mean ± SEM
FIGURE 2
FIGURE 2
In silico analysis of SORT1 gene expression on the survival of triple‐negative breast cancer (TNBC) patients. (A) TNBC patient survival curves were obtained using the Kaplan‐Meier plotter software (http://kmplot.com/analysis/) for patients expressing low (black curve) or high (red curve) levels of SORT1 (n = 161 cases) or (B) in TNBC patients with lymph node metastases (n = 72 cases)
FIGURE 3
FIGURE 3
SORT1‐mediated TH19P01 peptide uptake in MDA‐MB‐231 cells. Transient SORT1 gene silencing was performed as described in the Methods section and confirmed by (A) Western blotting and (B) densitometric analysis of SORT1 protein expression. Experiment was performed in triplicate and the densitometric data analyzed using a t‐test. (C) The binding (left panel) or uptake (right panel) of 200 nM Alexa488‐labeled TH19P01 was performed in control (siScrambled) or SORT1‐deficient (siSORT1) MDA‐MB‐231 cancer cells. The levels of fluorescence in the two categories of cells were measured in triplicate (binding and uptake n = 3). Affinity constants were extrapolated for (D) Alexa488‐labeled TH19P01 cell surface binding (KD), and inhibitory constants (Ki) for (E) uptake of 200 nM Alexa488‐labeled TH19P01 performed in the presence of increasing TH1902, TH19P01, or Neurotensin as indicated. Data are represented as mean ± SD and statistical analysis was performed using Student's unpaired t test (*P < .05, **P < .01)
FIGURE 4
FIGURE 4
Assessment of SORT1 expression requirement for TH19P01 peptide uptake in different cancer cell models. The uptake of 200 nM Alexa488‐labeled TH19P01 was evaluated in (A) human TNBC cells (MDA‐MB‐231), human colon cancer cells (MC38), and (B) murine ovarian cancer cells (ID8), and murine TNBC cells (4T1). This was performed in the absence (white bar) or presence (black bars) of excess unlabeled TH19P01 (50 µM), Neurotensin (NT, 10 µM) or Progranulin (PGRN, 1.5 nM). Data are represented as mean ± SD (ID8, n = 3; 4T1, n = 5; MDA‐MB‐231 and MC38, n = 2) and statistical analysis was performed using Dunnet's test (*P < .05, **P < .01). Inserts are representative immunodetections of Sortilin in whole cell lysates from each of the cell models tested
FIGURE 5
FIGURE 5
Schematic representation of docetaxel conjugation to the TH19P01 peptide. (A) Synthesis of TH1902 was performed as described in the Materials section. Briefly, DIEA was added dropwise to a solution of DoceSuOH and TBTU in DMSO in order to preactivate the DoceSuOH. The completion of preactivation was monitored by UPLC/MS, then a solution of TH19P01‐peptide in DMSO was added. The mixture was stirred at room temperature and the reaction was again monitored by UPLC/MS until completion. The reaction mixture was purified using a 30RPC resin column and an AKTA purifier system (10%‐80% ACN) to give TH19P01‐(SuDoce)2 or TH1902 as a white powder after lyophilization. (B) The purity (>95%) of the conjugate (peak 1) was evaluated by UPLC/MS, and (C) had an estimated m/z mass of 1852.91 (2+ charge)
FIGURE 6
FIGURE 6
In vitro anticancer properties of TH1902. A, A representative plot of the in vitro antiproliferative activities of TH19P01, docetaxel and TH1902 was determined from MTT assays in MDA‐MB‐231 as described in the Methods section. B, IC50 values were calculated using a log[inhibitor] vs response‐constant slope equation with GraphPad Prism software. IC50 values are presented as mean ± SD from human TNBC‐derived MDA‐MB‐231 and HCC‐70 cells. All assays were performed in quadruplicate (n = 3 experiments, except for TH1902 n = 4). C, A representative cell‐cycle phase distribution of MDA‐MB‐231 cells was determined based on cellular DNA content (propidium iodide staining) after a 2 h treatment with 4 µM docetaxel or 2 µM TH1902 then followed by a 22 h incubation in complete media alone. D, Quantification of MDA‐MB‐231 G2/M cell cycle arrest following docetaxel or TH1902 treatment. Data represent mean ± SD (n = 4)
FIGURE 7
FIGURE 7
Cell death induction by docetaxel and TH1902 in MDA‐MB‐231 cells. A, MDA‐MB‐231 cells were treated with vehicle, 10 µM docetaxel or 5 µM TH1902 for 24 h and cellular morphology was examined under a light microscope. B, MDA‐MB‐231 cells were treated for 5 or 24 h with 10 µM docetaxel (white bars), or with 5‐10 µM TH1902 (black bars). Cells were then harvested and the extent of apoptotic cell death determined by flow cytometry following staining with AnnexinV‐FITC and propidium iodide. Data are represented as mean ± SEM (n = 3, except for the 5 µM data where n = 7). C, TH1902‐mediated apoptosis in MDA‐MB‐231 cells was competed by an excess of free TH19P01 peptide (50 µM) or one of the SORT1 ligands neurotensin (NT, 10 µM) or progranulin (PGRN, 1 nM). Following 5 h of incubation, cells were stained with AnnexinV‐FITC/PI. All data are represented as mean ± SD (n = 3) and statistical analysis was performed using one‐way ANOVA with Dunnet's multiple comparison test (B and C) (*P < .05, **P < .01)
FIGURE 8
FIGURE 8
Effect of TH1902 on MDA‐MB‐231 microtubules and cell migration. A, MDA‐MB‐231 cells were treated with vehicle (DMSO), 2 µM docetaxel or 1 μM TH1902 for 24 h, fixed and immunostained with anti‐α‐tubulin antibody, DNA stained with DAPI, and then imaged using confocal fluorescence microscopy. Representative pictures from each condition are displayed. (B) Representative immunoblot showing the protein expression level of Bcl‐xL in MDA‐MB‐231 cells after treatment with docetaxel (100 nM) or TH1902 (50 nM) for 48 h. GAPDH was used as a loading control. Data are representative of three independent experiments. (C) Quantification of Bcl‐xL was performed by scanning densitometry. Data represent mean ± SD of arbitrary units and statistical analysis was performed using one‐way ANOVA with Dunnet's test compared to control, (*P < .05, n = 3). The impact of (D) docetaxel and (E) TH1902 on MDA‐MB‐231 cell migration was evaluated with and without siRNA‐mediated silencing of SORT1. Transfected cells were pretreated for 2 h with TH1902 (1 µM) or docetaxel (2 µM). Cells were harvested and their migratory potential assessed in real time as described in the Methods section. Data of normalized arbitrary units are represented as mean ± SD of two experiments performed in duplicate. Statistical analysis of data at 10 h was performed using one‐way ANOVA with Dunnet's multiple comparisons test (**P < .01)
FIGURE 9
FIGURE 9
Comparative efficacy of docetaxel and TH1902 on MDA‐MB‐231 xenograft tumor growth. A, Nude mice were subcutaneously implanted with MDA‐MB‐231‐Luc cells and then treated with vehicle, docetaxel or TH1902 as described in the Methods section. Intraperitoneal (i.p.) treatments are indicated by red arrows. B, Representative bioluminescence images were taken at 14 and 74 days post‐implantation of MDA‐MB‐231‐Luc cells into the mice. In the color bar, violet and red indicate the highest and lowest intensity of exposure, respectively. Results are expressed in terms of tumor luminescence intensity at day 0 post‐treatment for each mouse. C, Tumor burden was expressed as net luminescence intensity at days 14 and 74 post‐treatment. Data are represented as mean ± SEM (six mice/group). At day 74, all control mice were already sacrificed. Therefore, NA in this panel indicates that bioluminescence is not available (NA) for this group, whereas the bioluminescence in TH1902‐treated mice was below detectable levels (BDL)
FIGURE 10
FIGURE 10
Intravenous administration of TH1902 inhibits tumor growth in a MDA‐MB‐231 TNBC xenograft model. A, Nude mice were subcutaneously implanted with TNBC‐derived MDA‐MB‐231 cells and then treated with vehicle or with either docetaxel (15 mg/kg, three cycles) or TH1902 (35 mg/kg, five cycles) as described in the Methods section. Intravenous (i.v.) treatments are indicated by red arrows. B, Tumor volume progression measured as above for vehicle, docetaxel (3.75 mg/kg), or TH1902 (8.75 mg/kg) treatments. C, Representative bioluminescence images were taken at day 27 post‐implantation of MDA‐MB‐231‐Luc cells into the mice. In the color bar, violet and red indicate the highest and lowest intensity of exposure, respectively. D, Tumor burden was expressed as net luminescence intensity at day 27. Data are represented as mean ± SEM and statistical analysis was performed using one‐way ANOVA with Dunnet's test compared to control (A, n = 5 mice/group; B‐D, n = 6 mice/group, ***P < .001)
FIGURE 11
FIGURE 11
Intravenous administration of TH1902 inhibits tumor growth in a HCC‐70 TNBC xenograft model. A, Nude mice were subcutaneously implanted with TNBC‐derived HCC‐70 cells and then treated with vehicle or with either docetaxel (3.75 mg/kg, ¼ MTD) or TH1902 (8.75 mg/kg) as described in the Methods section. Intravenous treatments are indicated by red arrows. B, Tumor volume progression measured as above for vehicle, docetaxel (3.75 mg/kg), or TH1902 (8.75 mg/kg) treatments. Data are represented as mean ± SEM and statistical analysis was performed using one‐way ANOVA with Dunnet's test compared to control (vehicle, = 7; docetaxel and TH1902, n = 6 mice/group; **P < .01)
FIGURE 12
FIGURE 12
Absence of hematotoxicity of TH1902 in mice. A, Athymic nude mice were treated with docetaxel at MTD (15 mg/kg/wk, three cycles) or with an equivalent dose of TH1902 (up to six cycles). Blood was collected 4 days before the initiation of treatments and 4 days after the first, third and sixth treatments to assess the drug effects on neutrophil blood cell counts. Neutrophils count was not available (NA) for mice treated with docetaxel since treatment was stopped after three cycles. Data are represented as mean ± SEM and statistical analysis was performed using a t‐test against initial values observed at treatment 0 (n = 5 mice for treatments 0, 1 and 6; n = 2 mice for treatment 3; *P < .05, **P < .01). B, Body weight of mice was monitored with relation to the treatments. The number of mice with variations in body weight (>5% and 10%) after three or six treatments is presented (n = 5 mice/group)
FIGURE 13
FIGURE 13
Preliminary pharmacokinetics of TH1902 in mice. Normal CD‐1 mice were injected i.v. with TH1902 (50 mg/kg). Plasma samples for pharmacokinetic characterization were collected at the indicated times after i.v. bolus injection of TH1902. TH1902 and docetaxel concentrations were determined by UPLC/MS as described in the Methods section. Both TH1902 and free docetaxel plasmatic concentrations were plotted as a function of time. Cmax, termination half‐life and AUC were estimated using the PK solution software. Data are represented as mean ± SEM (n = 3 to 6 mice per time point)

References

    1. Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001;98:10869‐10874.
    1. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747‐752.
    1. Wolff AC, Hammond ME, Hicks DG, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013;31:3997‐4013.
    1. Rakha EA, Reis‐Filho JS, Ellis IO. Basal‐like breast cancer: a critical review. J Clin Oncol. 2008;26:2568‐2581.
    1. Parise CA, Caggiano V. Risk factors associated with the triple‐negative breast cancer subtype within four race/ethnicities. Breast Cancer Res Treat. 2017;163:151‐158.
    1. Newcomb PA, Storer BE, Longnecker MP, et al. Lactation and a reduced risk of premenopausal breast cancer. N Engl J Med. 1994;330:81‐87.
    1. McTiernan A, Thomas DB. Evidence for a protective effect of lactation on risk of breast cancer in young women. Results from a case‐control study. Am J Epidemiol. 1986;124:353‐358.
    1. Lowe CR, MacMahon B. Breast cancer and reproduction. Lancet. 1970;2:1137.
    1. Mirra AP, Cole P, MacMahon B. Breast cancer in an area of high parity: Sao Paulo, Brazil. Cancer Res. 1971;31:77‐83.
    1. Wetterskog D, Lopez‐Garcia MA, Lambros MB, et al. Adenoid cystic carcinomas constitute a genomically distinct subgroup of triple‐negative and basal‐like breast cancers. J Pathol. 2012;226:84‐96.
    1. Nunnery SE, Mayer IA, Balko JM. Triple‐negative breast cancer: breast tumors with an identity crisis. Cancer J. 2021;27(1):2‐7.
    1. Lehmann BD, Bauer JA, Chen X, et al. Identification of human triple‐negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121:2750‐2767.
    1. Lin NU, Vanderplas A, Hughes ME, et al. Clinicopathologic features, patterns of recurrence, and survival among women with triple‐negative breast cancer in the National Comprehensive Cancer Network. Cancer. 2012;118:5463‐5472.
    1. Anders CK, Carey LA. Biology, metastatic patterns, and treatment of patients with triple‐negative breast cancer. Clin Breast Cancer. 2009;9(Suppl 2):S73‐81.
    1. Cadoo KA, Fornier MN, Morris PG. Biological subtypes of breast cancer: current concepts and implications for recurrence patterns. Q J Nucl Med Mol Imaging. 2013;57:312‐321.
    1. Guarneri V, Dieci MV, Conte P. Relapsed triple‐negative breast cancer: challenges and treatment strategies. Drugs. 2013;73:1257‐1265.
    1. Montagna E, Bagnardi V, Rotmensz N, et al. Breast cancer subtypes and outcome after local and regional relapse. Ann Oncol. 2012;23:324‐331.
    1. de Ruijter TC, Veeck J, de Hoon JP, van Engeland M, Tjan‐Heijnen VC. Characteristics of triple‐negative breast cancer. J Cancer Res Clin Oncol. 2011;137:183‐192.
    1. Chacon RD, Costanzo MV. Triple‐negative breast cancer. Breast Cancer Res. 2010;12(Suppl 2):S3.
    1. Hudis CA, Gianni L. Triple‐negative breast cancer: an unmet medical need. Oncologist. 2011;16(Suppl 1):1‐11.
    1. Rapoport BL, Demetriou GS, Moodley SD, Benn CA. When and how do I use neoadjuvant chemotherapy for breast cancer? Curr Treat Options Oncol. 2014;15:86‐98.
    1. Lehmann BD, Pietenpol JA, Tan AR. Triple‐negative breast cancer: molecular subtypes and new targets for therapy. Am Soc Clin Oncol Educ Book. 2015;2015:e31‐e39. 10.14694/EdBook_AM.2015.35.e31
    1. Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced triple‐negative breast cancer: Phase Ib keynote‐012 study. J Clin Oncol. 2016;34(21):2460‐2467.
    1. Costa RLB, Gradishar WJ. Triple‐negative breast cancer: current practice and future directions. J Oncol Pract. 2017;13(5):301‐303.
    1. Han Y, Yu X, Li S, Tian Y, Liu C. New perspectives for resistance to PARP inhibitors in triple‐negative breast cancer. Front Oncol. 2020;10:578095.
    1. Gupta GK, Collier AL, Lee D, et al. Perspectives on triple‐negative breast cancer: current treatment strategies, unmet needs, and potential targets for future therapies. Cancers (Basel). 2020;12(9):2392.
    1. Marcusson EG, Horazdovsky BF, Cereghino JL, Gharakhanian E, Emr SD. The sorting receptor for yeast vacuolar carboxypeptidase Y is encoded by the VPS10 gene. Cell. 1994;77:579‐586.
    1. Vincent JP, Mazella J, Kitabgi P. Neurotensin and neurotensin receptors. Trends Pharmacol Sci. 1999;20:302‐309.
    1. Mazella J, Zsurger N, Navarro V, et al. The 100‐kDa neurotensin receptor is gp95/sortilin, a non‐G‐protein‐coupled receptor. J Biol Chem. 1998;273:6273‐6276.
    1. Hu F, Padukkavidana T, Vaegter CB, et al. Sortilin‐mediated endocytosis determines levels of the frontotemporal dementia protein, progranulin. Neuron. 2010;68:654‐667.
    1. Carlo AS, Gustafsen C, Mastrobuoni G, et al. The pro‐neurotrophin receptor sortilin is a major neuronal apolipoprotein E receptor for catabolism of amyloid‐beta peptide in the brain. J Neurosci. 2013;33:358‐370.
    1. Kjolby M, Nielsen MS, Petersen CM. Sortilin, encoded by the cardiovascular risk gene SORT1, and its suggested functions in cardiovascular disease. Curr Atheroscler Rep. 2015;17:496.
    1. Oh TJ, Ahn CH, Kim BR, et al. Circulating sortilin level as a potential biomarker for coronary atherosclerosis and diabetes mellitus. Cardiovasc Diabetol. 2017;16:92.
    1. Roselli S, Pundavela J, Demont Y, et al. Sortilin is associated with breast cancer aggressiveness and contributes to tumor cell adhesion and invasion. Oncotarget. 2015;6:10473‐10486.
    1. Ghaemimanesh F, Ahmadian G, Talebi S, et al. The effect of sortilin silencing on ovarian carcinoma cells. Avicenna J Med Biotechnol. 2014;6:169‐177.
    1. Truzzi F, Marconi A, Lotti R, et al. Neurotrophins and their receptors stimulate melanoma cell proliferation and migration. J Invest Dermatol. 2008;128:2031‐2040.
    1. Giorgi RR, Chile T, Bello AR, et al. Expression of neurotensin and its receptors in pituitary adenomas. J Neuroendocrinol. 2008;20:1052‐1057.
    1. Gao F, Griffin N, Faulkner S, et al. The membrane protein sortilin can be targeted to inhibit pancreatic cancer cell invasion. Am J Pathol. 2020;190:1931‐1942.
    1. Demeule M, Currie JC, Larocque A, et al. Increasing potency of anticancer drugs through sortilin receptor‐mediated cancer therapy: a new targeted approach for the treatment of ovarian cancer. Cancer Res. 2020;80(16 Supplement):1061.
    1. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1–2):55‐63.
    1. Schimpff RM, Gourmelen M, Scarcériaux V, Lhiaubet AM, Rostène W. Plasma neurotensin levels in humans: relation to hormone levels in diseases involving the hypothalamo‐pituitary‐thyroid axis. Eur J Endocrinol. 1995;133(5):534‐538.
    1. Meeter LH, Patzke H, Loewen G, et al. Progranulin levels in plasma and cerebrospinal fluid in granulin mutation carriers. Dement Geriatr Cogn Dis Extra. 2016;6(2):330‐340.
    1. Han TD, Shang DH, Tian Y. Docetaxel enhances apoptosis and G2/M cell cycle arrest by suppressing mitogen‐activated protein kinase signaling in human renal clear cell carcinoma. Genet Mol Res. 2016;5:15.
    1. Abal M, Andreu JM, Barasorain I. Taxanes: microtubule and centrosome targets, and cell cycle dependent mechanisms of action. Curr Cancer Drug Targets. 2003;3:193‐203.
    1. Wright N, Rida PCG, Aneja R. Tackling intra‐ and inter‐tumor heterogeneity to combat triple negative breast cancer. Front Biosci (Landmark Ed). 2017;22:1549‐1580.
    1. Baselga J, Gomez P, Greil R, et al. Randomized phase II study of the anti‐epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple‐negative breast cancer. J Clin Oncol. 2013;31:2586‐2592.
    1. Carey LA, Rugo HS, Marcom PK, et al. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple‐negative breast cancer. J Clin Oncol. 2012;30:2615‐2623.
    1. Baudino TA. Targeted cancer therapy: the next generation of cancer treatment. Curr Drug Discov Technol. 2015;12:3‐20.
    1. Perez‐Herrero E, Fernandez‐Medarde A. Advanced targeted therapies in cancer: drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm. 2015;93:52‐79.
    1. He R, Finan B, Mayer JP, DiMarchi RD. Peptide conjugates with small molecules designed to enhance efficacy and safety. Molecules. 2019;24(10):1855.
    1. Boohaker RJ, Lee MW, Vishnubhotla P, Perez JM, Khaled AR. The use of therapeutic peptides to target and to kill cancer cells. Curr Med Chem. 2012;19:3794‐3804.
    1. Marqus S, Pirogova E, Piva TJ. Evaluation of the use of therapeutic peptides for cancer treatment. J Biomed Sci. 2017;24:21.
    1. Adams GP, Schier R, McCall AM, et al. High affinity restricts the localization and tumor penetration of single‐chain fv antibody molecules. Cancer Res. 2001;61:4750‐4755.
    1. Rudnick SI, Lou J, Shaller CC, et al. Influence of affinity and antigen internalization on the uptake and penetration of Anti‐HER2 antibodies in solid tumors. Cancer Res. 2011;71:2250‐2259.
    1. Ali R, Sudhir RRaK. New Peptide Based Therapeutic Approaches. OMICS International; 2014.
    1. Hernandez‐Vargas H, Palacios J, Moreno‐Bueno G. Molecular profiling of docetaxel cytotoxicity in breast cancer cells: uncoupling of aberrant mitosis and apoptosis. Oncogene. 2007;26:2902‐2913.
    1. Nehme A, Varadarajan P, Sellakumar G, et al. Modulation of docetaxel‐induced apoptosis and cell cycle arrest by all‐ trans retinoic acid in prostate cancer cells. Br J Cancer. 2001;84:1571‐1576.
    1. Han TD, Shang DH, Tian Y. Docetaxel enhances apoptosis and G2/M cell cycle arrest by suppressing mitogen‐activated protein kinase signaling in human renal clear cell carcinoma. Genet Mol Res. 2016;15.
    1. Baguley BC. Multidrug resistance in cancer. Methods Mol Biol. 2010;596:1‐14.
    1. Mohammad RM, Muqbil I, Lowe L, et al. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol. 2015;35:S78‐S103.
    1. Krajewski S, Krajewska M, Shabaik A, et al. Immunohistochemical analysis of in vivo patterns of Bcl‐X expression. Cancer Res. 1994;54(21):5501‐5507.
    1. Hanada M, Aimé‐Sempé C, Sato T, Reed JC. Structure‐function analysis of Bcl‐2 protein. Identification of conserved domains important for homodimerization with Bcl‐2 and heterodimerization with Bax. J Biol Chem. 1995;270(20):11962‐11969.
    1. Olopade OI, Adeyanju MO, Safa AR, et al. Overexpression of BCL‐x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J Sci Am. 1997;3(4):230‐237.
    1. Simões‐Wüst AP, Olie RA, Gautschi O, et al. Bcl‐xl antisense treatment induces apoptosis in breast carcinoma cells. Int J Cancer. 2000;87(4):582‐590.
    1. Xu Z, Friess H, Solioz M, et al. Bcl‐xL antisense oligonucleotides induce apoptosis and increase sensitivity of pancreatic cancer cells to gemcitabine. Int J Cancer. 2001;94:268‐274.
    1. Ibrado AM, Liu L, Bhalla K. Bcl‐xL overexpression inhibits progression of molecular events leading to paclitaxel‐induced apoptosis of human acute myeloid leukemia HL‐60 cells. Cancer Res. 1997;57:1109‐1115.
    1. Wang Z, Goulet R 3rd, Stanton KJ, Sadaria M, Nakshatri H. Differential effect of anti‐apoptotic genes Bcl‐xL and c‐FLIP on sensitivity of MCF‐7 breast cancer cells to paclitaxel and docetaxel. Anticancer Res. 2005;25:2367‐2379.
    1. Lim LY, Vidnovic N, Ellisen LW, Leong CO. Mutant p53 mediates survival of breast cancer cells. Br J Cancer. 2009;101:1606‐1612.
    1. Morse DL, Gray H, Payne CM, Gillies RJ. Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol Cancer Ther. 2005;4:1495‐1504.
    1. Nieves‐Neira W, Pommier Y. Apoptotic response to camptothecin and 7‐hydroxystaurosporine (UCN‐01) in the 8 human breast cancer cell lines of the NCI Anticancer Drug Screen: multifactorial relationships with topoisomerase I, protein kinase C, Bcl‐2, p53, MDM‐2 and caspase pathways. Int J Cancer. 1999;82:396‐404.
    1. Ringel I, Horwitz SB. Studies with RP 56976 (taxotere): a semisynthetic analogue of taxol. J Natl Cancer Inst. 1991;83:288‐291.
    1. Shin DS, Ribas A. The evolution of checkpoint blockade as a cancer therapy: what's here, what's next? Curr Opin Immunol. 2015;33:23‐35.
    1. Emens LA, Kok M, Ojalvo LS. Targeting the programmed cell death‐1 pathway in breast and ovarian cancer. Curr Opin Obstet Gynecol. 2016;28:142‐147.
    1. Sun WY, Lee YK, Koo JS. Expression of PD‐L1 in triple‐negative breast cancer based on different immunohistochemical antibodies. J Transl Med. 2016;14:173.

Source: PubMed

3
Suscribir