Nomograms to Predict the Density of Tumor-Infiltrating Lymphocytes in Patients With High-Grade Serous Ovarian Cancer

Danian Dai, Lili Liu, He Huang, Shangqiu Chen, Bo Chen, Junya Cao, Xiaolin Luo, Feng Wang, Rongzhen Luo, Jihong Liu, Danian Dai, Lili Liu, He Huang, Shangqiu Chen, Bo Chen, Junya Cao, Xiaolin Luo, Feng Wang, Rongzhen Luo, Jihong Liu

Abstract

Background: Tumor-infiltrating lymphocytes (TILs) have important roles in predicting tumor therapeutic responses and progression, however, the method of evaluating TILs is complicated. We attempted to explore the association of TILs with clinicopathological characteristics and blood indicators, and to develop nomograms to predict the density of TILs in patients with high-grade serous ovarian cancer (HGSOC).

Methods: The clinical profiles of 197 consecutive postoperative HGSOC patients were retrospectively analyzed. Tumor tissues and matched normal fallopian tubes were immunostained for CD3+, CD8+, and CD4+ T cells on corresponding tissue microarrays and the numbers of TILs were counted using the NIH ImageJ software. The patients were classified into low- or high-density groups for each marker (CD3, CD4, CD8). The associations of the investigated TILs to clinicopathological characteristics and blood indicators were assessed and the related predictors for densities of TILs were used to develop nomograms; which were then further evaluated using the C-index, receiver operating characteristic (ROC) curves and calibration plots.

Results: Menopausal status, estrogen receptor (ER), Ki-67 index, white blood cell (WBC), platelets (PLT), lactate dehydrogenase (LDH), and carbohydrate antigen 153 (CA153) had significant association with densities of tumor-infiltrating CD3+, CD8+, or CD4+ T cells. The calibration curves of the CD3+ (C-index = 0.748), CD8+ (C-index = 0.683) and CD4+ TILs nomogram (C-index = 0.759) demonstrated excellent agreement between predictions and actual observations. ROC curves of internal validation indicated good discrimination for the CD8+ TILs nomogram [area under the curve (AUC) = 0.659, 95% CI 0.582-0.736] and encouraging performance for the CD3+ (AUC= 0.708, 95% CI 0.636-0.781) and CD4+ TILs nomogram (AUC = 0.730, 95% CI 0.659-0.801).

Conclusion: Menopausal status, ER, Ki-67 index, WBC, PLT, LDH, and CA153 could reflect the densities of T cells in the tumor microenvironment. Novel nomograms are conducive to monitor the immune status of patients with HGSOC and help doctors to formulate the appropriate treatment strategies.

Keywords: blood indicators; high-grade serous ovarian cancer; nomograms; tumor microenvironment; tumor-infiltrating lymphocytes.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Dai, Liu, Huang, Chen, Chen, Cao, Luo, Wang, Luo and Liu.

Figures

Figure 1
Figure 1
Expression of CD3+, CD8+, and CD4+ TILs in HGSOC tissues (Tu) and matched normal epithelium of the fallopian tubes (N), shown at 40× magnification with inset (400×). (A–C) Representative images of CD3+, CD8+, and CD4+ TILs in the normal fallopian tube tissues. (D, G) Representative images of low-density and high-density of CD3+ TILs in the HGSOC tissues. (E, H) Representative images of low-density and high-density CD8+ TILs in the HGSOC tissues. (F, I) Representative images of low-density and high-density of CD4+ TILs in the HGSOC tissues. (J) Box plots of CD3+, CD8+, and CD4+ TILs per mm2 in tumor tissues (n=197) or normal epithelium of the fallopian tubes (n=173). Quantitative data are presented as mean ± SEM. TILs, tumor-infiltrating lymphocytes; HGSOC, high-grade serous ovarian cancer.
Figure 2
Figure 2
Immunohistochemical staining of H&E, ER, Ki-67, and p53 in HGSOC tissues (Tu) and matched normal fallopian tubes tissue (N), shown at 40× magnification with inset (400×). (A, B) Representative images of H&E in the matched normal epithelium of the fallopian tubes (N) and HGSOC tissues (Tu). (C, D) Representative images of negative and positive ER in HGSOC tissues are presented. (E, F) Representative images of high and low Ki-67 index in HGSOC tissues are shown (G, H) Representative images of mutant type and wild type p53 staining in HGSOC tissues are presented. TILs, tumor-infiltrating lymphocytes; HGSOC, high-grade serous ovarian cancer; H&E, Hematoxylin and Eosin.
Figure 3
Figure 3
Correlation between various blood indicators and the density of TILs. (A) The density of CD3+ TILs showed a positive correlation with the level of serum LDH, with a coefficient of 0.153 (p = 0.031). (B) The density of CD3+ TILs was negatively related to the PLT, with a coefficient of −0.186 (P = 0.009). (C) The density of CD3+ TILs showed a positive correlation the level of serum CA153, with a coefficient of 0.168 (P = 0.019). (D) The density of CD8+ TILs showed a tendency that positively correlated with the level of serum LDH, with a coefficient of 0.111 (P = 0.120). (E) The density of CD8+ TILs showed a tendency that positively related to the serum CA153, with a coefficient of 0.133 (P = 0.065). (F) The density of CD4+ TILs showed a tendency that negatively correlated with the number of WBC in the blood, with a coefficient of -0.126 (P = 0.079). (G) The density of CD4+ TILs was positively related to the CA153, with a coefficient of 0.207 (P = 0.004). TILs, tumor-infiltrating lymphocytes; WBC, white blood cells; LDH, lactate dehydrogenase; PLT, platelet; CA153, carbohydrate antigen 153.
Figure 4
Figure 4
Nomograms, calibration curves, and ROC curves analysis for predicting density of TILs in patients with high-grade serous ovarian cancer. (A) The CD3+ TILs prediction nomogram. (B) Calibration curves for predicting density of CD3+ TILs. (C) ROC curves of CD3+ TILs prediction nomogram in the internal testing set. (D) The CD8+ TILs prediction nomogram. (E) Calibration curves for predicting density of CD8+ TILs. (F) ROC curves of CD8+ TILs prediction nomogram in the internal testing set. (G) The CD4+ TILs prediction nomogram. (H) Calibration curves for predicting density of CD4+ TILs. (I) ROC curves of CD4+ TILs prediction nomogram in the internal testing set. All the points assigned on the top point scale for each factor are summed together to generate a total point score. The total point score is projected on the bottom scales to determine the probability of high density for tumor-infiltrating T cells in an individual. The nomogram-predicted frequency of high T cell density is plotted on the x-axis, and the actual observed frequency of high T cell density is plotted on the y-axis. The AUC was calculated, and its 95% CI was estimated by bootstrapping. TILs, tumor-infiltrating lymphocytes; ROC, receiver operating characteristic; 95% CI, 95% confidence interval.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA: Cancer J Clin (2019) 69(1):7–34. 10.3322/caac.21551
    1. Bowtell DD, Böhm S, Ahmed AA, Aspuria P-J, Bast RC, Jr., Beral V, et al. . Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer. Nat Rev Cancer (2015) 15(11):668–79. 10.1038/nrc4019
    1. Prat J. Oncology FCoG. FIGO’s staging classification for cancer of the ovary, fallopian tube, and peritoneum: abridged republication. J Gynecol Oncol (2015) 26(2):87–9. 10.3802/jgo.2015.26.2.87
    1. Miyamoto M, Takano M, Goto T, Kato M, Sasaki N, Tsuda H, et al. . Clear cell histology as a poor prognostic factor for advanced epithelial ovarian cancer: a single institutional case series through central pathologic review. J Gynecol Oncol (2013) 24(1):37–43. 10.3802/jgo.2013.24.1.37
    1. Konecny GE, Wang C, Hamidi H, Winterhoff B, Kalli KR, Dering J, et al. . Prognostic and therapeutic relevance of molecular subtypes in high-grade serous ovarian cancer. JNCI: J Natl Cancer Inst (2014) 106(10). 10.1093/jnci/dju249
    1. Bolton KL, Chenevix-Trench G, Goh C, Sadetzki S, Ramus SJ, Karlan BY, et al. . Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA (2012) 307(4):382–9. 10.1001/jama.2012.20
    1. Pal MK, Jaiswar SP, Dwivedi VN, Tripathi AK, Dwivedi A, Sankhwar P. MicroRNA: a new and promising potential biomarker for diagnosis and prognosis of ovarian cancer. Cancer Biol Med (2015) 12(4):328. 10.7497/j.issn.2095-3941.2015.0024
    1. Hwang W-T, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol (2012) 124(2):192–8. 10.1016/j.ygyno.2011.09.039
    1. Santoiemma PP, Powell DJ., Jr. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther (2015) 16(6):807–20. 10.1080/15384047.2015.1040960
    1. Eggermont A, Robert C, Soria JC, Zitvogel L. Harnessing the immune system to provide long-term survival in patients with melanoma and other solid tumors. Oncoimmunology (2014) 3(1):e27560. 10.4161/onci.27560
    1. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases—elimination, equilibrium and escape. Curr Opin Immunol (2014) 27:16–25. 10.1016/j.coi.2014.01.004
    1. Fu Q, Chen N, Ge C, Li R, Li Z, Zeng B, et al. . Prognostic value of tumor-infiltrating lymphocytes in melanoma: a systematic review and meta-analysis. Oncoimmunology (2019) 8(7):1593806. 10.1080/2162402X.2019.1593806
    1. Idos GE, Kwok J, Bonthala N, Kysh L, Gruber SB, Qu C. The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis. Sci Rep (2020) 10(1):3360. 10.1038/s41598-020-60255-4
    1. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. . Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci (2005) 102(51):18538–43. 10.1073/pnas.0509182102
    1. Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, et al. . Assessing tumor infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in melanoma, gastrointestinal tract carcinomas, non-small cell lung carcinoma and mesothelioma, endometrial and ovarian carcinomas, squamous cell carcinoma of the head and neck, genitourinary carcinomas, and primary brain tumors. Adv Anat Pathol (2017) 24(6):311. 10.1097/PAP.0000000000000161
    1. Jeschke J, Bizet M, Desmedt C, Calonne E, Dedeurwaerder S, Garaud S, et al. . DNA methylation–based immune response signature improves patient diagnosis in multiple cancers. J Clin Invest (2017) 127(8):3090–102. 10.1172/JCI91095
    1. McGrail DJ, Federico L, Li Y, Dai H, Lu Y, Mills GB, et al. . Multi-omics analysis reveals neoantigen-independent immune cell infiltration in copy-number driven cancers. Nat Commun (2018) 9(1):1–13. 10.1038/s41467-018-03730-x
    1. Deeba F, Khatun S, Alam M, Shahida S. Serum LDH and CA-125: Markers for Diagnosis of Ovarian Malignancy. Mymensingh Med J: MMJ (2015) 24(2):334–40.
    1. Williams KA, Labidi-Galy SI, Terry KL, Vitonis AF, Welch WR, Goodman A, et al. . Prognostic significance and predictors of the neutrophil-to-lymphocyte ratio in ovarian cancer. Gynecol Oncol (2014) 132(3):542–50. 10.1016/j.ygyno.2014.01.026
    1. Asher V, Lee J, Innamaa A, Bali A. Preoperative platelet lymphocyte ratio as an independent prognostic marker in ovarian cancer. Clin Trans Oncol (2011) 13(7):499. 10.1007/s12094-011-0687-9
    1. Eo WK, Chang HJ, Kwon SH, Koh SB, Kim YO, Ji YI, et al. . The lymphocyte-monocyte ratio predicts patient survival and aggressiveness of ovarian cancer. J Cancer (2016) 7(3):289. 10.7150/jca.13432
    1. Høgdall EV, Christensen L, Kjaer SK, Blaakaer J, Kjærbye-Thygesen A, Gayther S, et al. . CA125 expression pattern, prognosis and correlation with serum CA125 in ovarian tumor patients: From The Danish “MALOVA” ovarian cancer study. Gynecol Oncol (2007) 104(3):508–15. 10.1016/j.ygyno.2006.09.028
    1. Feng X, Juan X. Diagnostic value of combined detection of CA125, CA153, CA199 and CEA in epithelial ovarian carcinoma. Int J Lab Med (2014) 14:1846–7,1850.
    1. Balachandran VP, Gonen M, Smith JJ, DeMatteo RP. Nomograms in oncology: more than meets the eye. Lancet Oncol (2015) 16(4):e173–e80. 10.1016/S1470-2045(14)71116-7
    1. Shi H, Jiang Y, Cao H, Zhu H, Chen B, Ji W. Nomogram based on systemic immune-inflammation index to predict overall survival in gastric cancer patients. Dis Markers (2018) 2018:1787424. 10.1155/2018/1787424
    1. Diao X, Cai J, Zheng J, Kong J, Wu S, Yu H, et al. . Association of chromosome 7 aneuploidy measured by fluorescence in situ hybridization assay with muscular invasion in bladder cancer. Cancer Commun (Lond) (2020) 40(4):167–80. 10.1002/cac2.12017
    1. Dai D, Chen B, Tang H, Wang B, Zhao Z, Xie X, et al. . Nomograms for predicting the prognostic value of pre-therapeutic CA15-3 and CEA serum levels in TNBC patients. PLoS One (2016) 11(8):e0161902. 10.1371/journal.pone.0161902
    1. Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, et al. . Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol (2014) 232(2):199–209. 10.1002/path.4287
    1. Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. . American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer (unabridged version). Arch Pathol Lab Med (2010) 134(7):e48–72.
    1. Hosmer DW, Jr, Lemeshow S, Sturdivant RX. Applied logistic regression. New York: John Wiley & Sons; (2013). 10.1002/9781118548387
    1. Kramer AA, Zimmerman JE. Assessing the calibration of mortality benchmarks in critical care: The Hosmer-Lemeshow test revisited. Crit Care Med (2007) 35(9):2052–6. 10.1097/01.CCM.0000275267.64078.B0
    1. Grossman CJ. Regulation of the immune system by sex steroids. Endocr Rev (1984) 5(3):435–55. 10.1210/edrv-5-3-435
    1. Yang J-H, Chen C-D, Wu M-Y, Chao K-H, Yang Y-S, Ho H-N. Hormone replacement therapy reverses the decrease in natural killer cytotoxicity but does not reverse the decreases in the T-cell subpopulation or interferon-gamma production in postmenopausal women. Fertil Steril (2000) 74(2):261–7. 10.1016/S0015-0282(00)00622-1
    1. Kumru S, Godekmerdan A, Yılmaz B. Immune effects of surgical menopause and estrogen replacement therapy in peri-menopausal women. J Reprod Immunol (2004) 63(1):31–8. 10.1016/j.jri.2004.02.001
    1. Li L, Wu J, Pu D, Zhao Y, Wan C, Sun L, et al. . Factors associated with the age of natural menopause and menopausal symptoms in Chinese women. Maturitas (2012) 73(4):354–60. 10.1016/j.maturitas.2012.09.008
    1. Tomsová M, Melichar B, Sedláková I, Steiner I. Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol (2008) 108(2):415–20. 10.1016/j.ygyno.2007.10.016
    1. Kim ST, Jeong H, Woo OH, Seo JH, Kim A, Lee ES, et al. . Tumor-infiltrating lymphocytes, tumor characteristics, and recurrence in patients with early breast cancer. Am J Clin Oncol (2013) 36(3):224–31. 10.1097/COC.0b013e3182467d90
    1. Macciò A, Madeddu C, Massa D, Astara G, Farci D, Melis GB, et al. . Interleukin-6 and leptin as markers of energy metabolicchanges in advanced ovarian cancer patients. J Cell Mol Med (2009) 13(9b):3951–9. 10.1111/j.1582-4934.2008.00408.x
    1. Macciò A, Madeddu C. Inflammation and ovarian cancer. Cytokine (2012) 58(2):133–47. 10.1016/j.cyto.2012.01.015
    1. Li G, Gao J, Tao Y-L, Xu B-Q, Tu Z-W, Liu Z-G, et al. . Increased pretreatment levels of serum LDH and ALP as poor prognostic factors for nasopharyngeal carcinoma. Chin J Cancer (2012) 31(4):197. 10.5732/cjc.011.10283
    1. Thonsri U, Seubwai W, Waraasawapati S, Sawanyawisuth K, Vaeteewoottacharn K, Boonmars T, et al. . Overexpression of lactate dehydrogenase A in cholangiocarcinoma is correlated with poor prognosis. Histol Histopathol (2017) 32(5):503–10. 10.14670/HH-11-819
    1. Pinto MP, Balmaceda C, Bravo ML, Kato S, Villarroel A, Owen GI, et al. . Patient inflammatory status and CD4+/CD8+ intraepithelial tumor lymphocyte infiltration are predictors of outcomes in high-grade serous ovarian cancer. Gynecol Oncol (2018) 151(1):10–7. 10.1016/j.ygyno.2018.07.025
    1. Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, et al. . LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab (2016) 24(5):657–71. 10.1016/j.cmet.2016.08.011
    1. Stone RL, Nick AM, McNeish IA, Balkwill F, Han HD, Bottsford-Miller J, et al. . Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med (2012) 366(7):610–8. 10.1056/NEJMoa1110352
    1. Rachidi S, Metelli A, Riesenberg B, Wu BX, Nelson MH, Fugle CW, et al. . Platelets subvert T cell immunity against cancer via GARP-TGFβ axis. Sci Immunol (2017) 2(11):eaai7911. 10.1126/sciimmunol.aai7911
    1. Li H, Manwani B, Leng SX. Frailty, inflammation, and immunity. Aging Dis (2011) 2(6):466–73.
    1. Lewkowicz P, Lewkowicz N, Sasiak A, Tchórzewski H. Lipopolysaccharide-activated CD4+ CD25+ T regulatory cells inhibit neutrophil function and promote their apoptosis and death. J Immunol (2006) 177(10):7155–63. 10.4049/jimmunol.177.10.7155
    1. Deng J, Wang L, Chen H, Li L, Ma Y, Ni J, et al. . The role of tumour-associated MUC1 in epithelial ovarian cancer metastasis and progression. Cancer Metastasis Rev (2013) 32(3-4):535–51. 10.1007/s10555-013-9423-y
    1. Feng H, Ghazizadeh M, Konishi H, Araki T. Expression of MUC1 and MUC2 mucin gene products in human ovarian carcinomas. Jpn J Clin Oncol (2002) 32(12):525–9. 10.1093/jjco/hyf111
    1. McDermott KM, Crocker PR, Harris A, Burdick MD, Hinoda Y, Hayashi T, et al. . Overexpression of MUC1 reconfigures the binding properties of tumor cells. Int J Cancer (2001) 94(6):783–91. 10.1002/ijc.1554
    1. Chan AK, Lockhart DC, von Bernstorff W, Spanjaard RA, JooT HG, Eberlein J, et al. . Soluble MUC1 secreted by human epithelial cancer cells mediates immune suppression by blocking T-cell activation. Int J Cancer (1999) 82(5):721–6. 10.1002/(SICI)1097-0215(19990827)82:5<721::AID-IJC16>;2-N
    1. Budiu R, Elishaev E, Brozick J, Lee M, Edwards R, Kalinski P, et al. . Immunobiology of human mucin 1 in a preclinical ovarian tumor model. Oncogene (2013) 32(32):3664–75. 10.1038/onc.2012.397
    1. Wright AA, Bohlke K, Armstrong DK, Bookman MA, Cliby WA, Coleman RL, et al. . Neoadjuvant chemotherapy for newly diagnosed, advanced ovarian cancer: Society of Gynecologic Oncology and American Society of Clinical Oncology clinical practice guideline. Gynecol Oncol (2016) 143(1):3–15. 10.1016/j.ygyno.2016.05.022
    1. Narod S, Sopik V. Neoadjuvant chemotherapy for advanced-stage ovarian cancer: Are the ASCO and SGO recommendations warranted? Gynecol Oncol (2017) 144(2):238–40. 10.1016/j.ygyno.2016.11.046
    1. Mao Y, Qu Q, Zhang Y, Liu J, Xiaosong C, Shen K. The value of tumor infiltrating lymphocytes (TILs) for predicting response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis. PLoS One (2014) 9(12):e115103. 10.1371/journal.pone.0115103
    1. Lo CS, Sanii S, Kroeger DR, Milne K, Talhouk A, Chiu DS, et al. . Neoadjuvant chemotherapy of ovarian cancer results in three patterns of tumor-infiltrating lymphocyte response with distinct implications for immunotherapy. Clin Cancer Res (2017) 23(4):925–34. 10.1158/1078-0432.CCR-16-1433
    1. Khairallah A, Genestie C, Auguste A, Leary A. Impact of neoadjuvant chemotherapy on the immune microenvironment in advanced epithelial ovarian cancer: Prognostic and therapeutic implications. Int J Cancer (2018) 143(1):8–15. 10.1002/ijc.31200
    1. Kowanetz M, Zou W, Gettinger S, Koeppen H, Kockx M, Schmid P, et al. . Differential regulation of PD-L1 expression by immune and tumor cells in NSCLC and the response to treatment with atezolizumab (anti-PD-L1). Proc Natl Acad Sci U S A (2018) 115(43):E10119–26. 10.1073/pnas.1802166115
    1. Wu X, Feng Q, Wang Y, Shi J, Ge H, Di W. The immunologic aspects in advanced ovarian cancer patients treated with paclitaxel and carboplatin chemotherapy. Cancer Immunol Immunother: CII (2010) 59(2):279–91. 10.1007/s00262-009-0749-9
    1. Proietti E, Moschella F, Capone I, Belardelli F. Exploitation of the propulsive force of chemotherapy for improving the response to cancer immunotherapy. Mol Oncol (2012) 6(1):1–14. 10.1016/j.molonc.2011.11.005
    1. Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. . The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol (2015) 26(2):259–71. 10.1093/annonc/mdu450
    1. Tan WCC, Nerurkar SN, Cai HY, Ng HHM, Wu D, Wee YTF, et al. . Overview of multiplex immunohistochemistry/immunofluorescence techniques in the era of cancer immunotherapy. Cancer Commun (Lond) (2020) 40(4):135–53. 10.1002/cac2.12023
    1. Rutten M, Boldingh J, Schuit E, Trum H, Van Driel W, Mol B, et al. . Development and internal validation of a prognostic model for survival after debulking surgery for epithelial ovarian cancer. Gynecol Oncol (2014) 135(1):13–8. 10.1016/j.ygyno.2014.07.099
    1. Shim S-H, Lee SJ, Kim S-O, Kim S-N, Kim D-Y, Lee JJ, et al. . Nomogram for predicting incomplete cytoreduction in advanced ovarian cancer patients. Gynecol Oncol (2015) 136(1):30–6. 10.1016/j.ygyno.2014.11.004
    1. Hwang HW, Jung H, Hyeon J, Park YH, Ahn JS, Im Y-H, et al. . A nomogram to predict pathologic complete response (pCR) and the value of tumor-infiltrating lymphocytes (TILs) for prediction of response to neoadjuvant chemotherapy (NAC) in breast cancer patients. Breast Cancer Res Treat (2019) 173(2):255–66. 10.1007/s10549-018-4981-x

Source: PubMed

3
Abonnere