Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2020 Feb 13;69(5):799–811. doi: 10.1007/s00262-020-02512-z

Co-expression of HLA-I loci improved prognostication in HER2+ breast cancers

Julia Y Tsang 1, Chun-Sing Ho 1,3, Yun-Bi Ni 1, Yan Shao 1, Ivan K Poon 1, Siu-Ki Chan 2, Sai-Yin Cheung 3, Ka-Ho Shea 3, Monalyn Marabi 4, Gary M Tse 1,
PMCID: PMC11027840  PMID: 32055918

Abstract

The underlying basis for cancer immune evasion is important for effective immunotherapy and prognosis in breast cancers. Human leucocyte antigens (HLA)-I comprising three classical antigens (HLA-A, -B and -C) is mandatory for anti-tumor immunity. Its loss occurred frequently in many cancers resulting in effective immune evasion. Most studies examined HLA-I as a whole. Alterations in specific locus could have different clinical ramifications. Hence, we evaluated the expression of the three HLA-I loci in a large cohort of breast cancers. Low expression of HLA-A, -B and -C were found in 71.1%, 66.3%, and 60.2% of the cases. Low and high expression in all loci was found in 48.3% and 17.9% of the cases respectively. The remaining showed high expression in one or two loci. Cases with all HLA high expression (all HLA high) was frequent in the ER-HER2- (27.4%) and ER-HER2+ (23.1%) cases and was associated with characteristic pathologic features related to these tumor (higher grade, necrosis, high tumor infiltrating lymphocyte (TIL), pT stage, low hormonal receptor, high basal marker expression) (p ≤ 0.019). Interestingly, in HER2+ cancers, only cases with all HLA high and high TIL showed significantly better survival. In node positive cancers, concordant high HLA expression in primary tumors and nodal metastases was favorable prognostically (DFS: HR = 0.741, p < 0.001; BCSS: HR = 0.699, p = 0.003). The data suggested an important clinical value of a combined analysis on the co-expression HLA-I status in both primary and metastatic tumors. This could be a potential additional key component to be incorporated into TIL evaluation for improved prognostication.

Electronic supplementary material

The online version of this article (10.1007/s00262-020-02512-z) contains supplementary material, which is available to authorized users.

Keywords: Breast cancer, HLA loci, Immunohistochemistry, Survival, HER2+

Introduction

The studies on tumor immunology have revolutionized our understanding of cancer pathogenesis and treatment. Immunotherapy for cancer management is now widely adopted in various cancer types. Durable responses and improved survival had been observed in patients treated with immune checkpoint blockade across a wide range of cancers [1]. Currently immunotherapy has been approved for melanoma, non-small-cell lung cancer, head and neck cancer, renal cell carcinoma and bladder cancer [1], and very recently in advanced triple negative breast cancer [2]. Despite that, biomarker development for accurate prediction of treatment response is less successful as cancer immune interaction is multifactorial. A better understanding of anti-tumor immunity is important to decipher the underlying mechanism of cancer immunosuppression and aid in biomarker development.

Tumor-associated antigens presenting by major histocompatibility complex (MHC) class I molecules to cytotoxic T cells is fundamental to anti-tumor immunity. MHC class I molecules are encoded by genes located within the MHC region on chromosome 6 and comprise three classical human leukocyte antigens (HLA-I), namely HLA-A, -B and-C in humans. In normal cells, six different HLA alleles, two from each locus, are expressed. These HLA alleles form a tri-molecular complex with β2-microglobulin and peptide antigen on the cell surface for antigen presentation. Loss of HLA-I expression is a frequent event in different cancers for immune evasion, allowing tumor dissemination and metastasis [3]. Several mechanisms of HLA loss have been proposed, leading to different patterns of HLA-I expression on cell surface. Depending on whether the genomic deletion and epigenetic regulation affect the entire HLA-I region, single locus or specific allele, HLA-I can be totally or partially absent from tumor cells. They can be downregulated by alterations in its regulatory signaling pathways and defects in the assembly and stability of HLA complex, such as downregulation of antigen processing machinery and β2-microglobulin. These can lead to loss of expression in all HLA-I loci [4].

In breast cancers, downregulation of HLA-I expression is common, being observed in 47–85% of primary tumors [513]. High HLA expression was consistently associated with higher tumor grade [5, 8, 9, 11], and variably with hormonal receptor negativity [5], HER2 negativity [13], lymphovascular invasion [9], and tumor infiltrating lymphocytes (TIL) [9]. Its relationship with outcome was also variable [5, 6, 8, 11, 13]. Most studies used antibodies that recognized the non-polymorphic region of HLA-I, thus did not differentiate the expression of the different HLA loci [513]. It is possible that alterations in specific locus could be observed with different clinical ramifications. In fact, in colon carcinoma, only alterations in HLA-A and C had prognostic relevance [14]. In endometrial cancer, loss of HLA-B and C, but not HLA-A, was associated with deficient mismatch repair [15].

In this study, we examined HLA-I expression in breast cancer using antibodies reactive to different HLA-I loci. The expression of the different HLAs and their co-expression status were assessed with various clinico-pathologic features, biomarker expression and patients’ survival. In addition, analysis was performed on paired primary tumors and corresponding nodal metastases to examine the relationship of HLA expression with tumor dissemination.

Materials and methods

Patients data

All consecutive cases diagnosed with breast cancer over a period of 4 (2002–2005), 7 (2003–2009) and 4 (2003–2006) years in three of the involved institutions were included. Patients’ demographic data (age) histopathologic parameters (tumor size, lymph node involvement, pN stage and pT stage) and outcome data were retrieved from the medical records. Disease-free survival (DFS) time was calculated from the date of the surgery to the date of the first relapse or death. Breast cancer specific survival (BCSS) time was calculated from the date of surgery to the date of the last follow-up, or death due to breast cancer. All the specimens were fixed in 10% buffered formalin and embedded in paraffin. Archival hematoxylin and eosin (H&E) stained slides for each case were reviewed to confirm the diagnosis (WHO criteria) and grade (Bloom and Richardson grading). Additional histologic features [including lympho-vascular invasion (LVI), phenotypic apocrine feature, necrosis, stromal TIL, and extensive in situ components (EIC)] were assessed as previously reported [16]. The study was approved by Joint Chinese University of Hong Kong—New Territories East Cluster clinical research ethics committee. Tissue from patients was acquired with informed consent in accordance with local institutional review and the Declaration of Helsinki.

Tissue microarray (TMA) construction and immunohistochemistry

TMA were prepared as previously described [16]. Briefly, representative areas of each tumor were selected and 0.6 mm core in duplicate were taken from each case for TMA construction. The presence of tumor was confirmed on H&E stained TMA sections. Immunohistochemical (IHC) staining was carried out on TMA sections with the selected antibodies using Ultraview Universal DAB Detection Kit (Ventana, Arizona, USA) after deparaffinization, rehydration and antigen retrieval of the slides. All slides were counterstained with hematoxylin. The IHC staining was evaluated on the basis of staining intensity (graded from 0 to 3) and the percentage of positively stained cells in the corresponding cellular location according to different antibodies. The interpretation of IHC results were carried out blindly by two of the authors without any clinical information and the staining results of other markers. Any discrepancies were resolved by discussion to reach a consensus. For HLA-A, -B and -C analysis, the evaluation was based mainly on membranous staining. An immunoscore was obtained by multiplying the intensity score with percentage of stained cells, giving a range of 0–300. The cases were divided into high and low expressing groups using the respective mean immunoscore as cutoff. (representative staining of the HLAs were shown in supplementary figure S1.) IHC results for other markers [including estrogen receptor (ER), progesterone receptor (PR), Ki67, human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), c-kit, P63, Cytokeratin (CK) 5/6, CK14, androgen receptor (AR), vimentin and p-cadherin, phosphorylated AKT (pAKT)] were retrieved from our database. Details of staining and assessment of all markers involved in the study were shown in Supplementary Table S1.

Statistical analysis

SPSS for Windows version 23.0 (SPSS Inc., Chicago, IL) was used for all statistical analyses. Chi-square analysis or Fisher’s exact test were used to test for the association between categorical variables. Mann–Whitney U test was used for analyzing the differences in continuous variables. Paired comparison between lymph node (LN) and primary tumor was performed using Wilcoxon signed rank test. Survival data were analyzed using the Kaplan–Meier method and group differences in survival time were investigated by a log-rank test. Multivariate Cox proportional hazards model with backward Wald model was used to identify variables that were independently associated with survival. All statistical tests were two-sided, and p value of < 0.05 was considered statistically significant.

Results

In total, 1190 primary invasive breast cancers and 314 paired metastatic lymph nodes were included in this cohort. The mean patients’ age at diagnosis was 54.4 ± 12.7 years (range 22–97 years) and the mean tumor size was 2.69 ± 1.55 cm (range 0.2–13.0 cm). Of the 1190 cases, 175 (14.7%), 477 (40.1%), and 538 (45.2%) were of grade I, II, and III respectively. ER, PR, and HER2 were positive in 70.3% (832/1183), 66.2% (780/1179), 19.0% (225/1184) of the cases respectively. High HLA-A, B and C expression was found in 27.8% (311/1120), 33.3% (380/1141) and 40.1% (452/1126) respectively. Among the 1054 cases with data on all three HLAs, 698 (66.2%) showed concordant expression (including 509 (48.3%) cases with low expression for all three markers (all low) and 189 (17.9%) cases showed all HLA high (all high) expression). The remainder (33.8%; 356/1054) expressed mixed levels of the three HLAs (mixed low/high). (Details on the combined expression of the HLA-A, B and C were shown at supplementary table S2).

Correlation with clinico-pathologic features, biomarkers, and breast cancer subtypes

All three HLAs showed significant correlation with each other (p < 0.001). High HLA-A expression was associated with higher grade (p < 0.001), absence of EIC (p = 0.007), presence of necrosis (p < 0.001) and high TIL level (p < 0.001). Differential level of HLA-A was found in breast cancer molecular subtypes defined by ER/HER expression (p < 0.001), with the lowest rate in ER + HER2- cancers. Similar associations were also shown for high HLA-B (p ≤ 0.013) and HLA-C (p ≤ 0.004) expression. In addition, high HLA-A (p = 0.003) and HLA-B (p = 0.001) expression was associated with higher pT stage. High HLA-A expression was associated with younger age (p = 0.015); high HLA-B expression was associated with phenotypic apocrine feature (p = 0.045); and high HLA-C expression was associated with FF (p = 0.002) and lower pN stage (p = 0.033). None showed correlation with LVI (Table 1). Among the biomarkers, all three HLAs correlated negatively with ER (p ≤ 0.002) and PR (p ≤ 0.007); but positively with Ki67 (p ≤ 0.001), CK5/6 (p ≤ 0.011), CK14 (p ≤ 0.048), vimentin (p ≤ 0.022), p-cadherin (p ≤ 0.008) and pAKT (p ≤ 0.037). HLA-A and HLA-C also showed positive association with HER2 (p ≤ 0.015), EGFR (p ≤ 0.006) and p63 (p ≤ 0.001). HLA-A was also associated with c-kit positivity (p = 0.011) while HLA-B was associated negatively with AR expression (p = 0.036) (Table 1).

Table 1.

Clinico-pathological association and biomarker profile of HLA-A, B and C

HLA-A HLA-B HLA-C
Low High Total p value Low high Total p value Low High Total p value
Clinico-pathological features
 Grade
  1 140 18 158 < 0.001 132 31 163 < 0.001 115 44 159 < 0.001
  2 352 96 448 330 129 459 289 156 445
  3 317 197 514 299 220 519 270 252 522
 LVI
  Absent 563 208 771 0.314 531 255 786 0.178 457 312 769 0.864
  Present 207 89 296 191 111 302 180 120 300
 Apocrine
  Absent 740 280 1020 0.482 697 335 1032 0.045 613 403 1016 0.300
  Present 65 29 94 57 42 99 55 45 100
 FF
  Absent 585 239 824 0.137 561 279 840 0.635 475 357 832 0.002
  Present 208 67 275 182 97 279 183 88 271 -0.092
 EIC
  Absent 631 264 895 0.007 586 319 905 0.013 515 377 892 0.003
  Present 169 43 212 164 59 223 151 69 220
 Necrosis
  Absent 649 216 865 < 0.001 606 275 881 < 0.001 532 332 864 0.004
  Present 129 87 216 120 100 220 113 109 222
 LI
  Low 548 138 686 < 0.001 504 192 696 < 0.001 446 238 684 < 0.001
  High 82 89 171 80 95 175 68 102 170
 pT
  1 351 93 444 0.003 334 118 452 0.001 283 161 444 0.119
  2 388 194 582 366 228 594 332 258 590
  3 45 14 59 44 18 62 38 20 58
  4 16 7 23 12 10 22 12 10 22
 pN
  0 398 139 537 0.203 375 179 554 0.331 310 234 544 0.033
  1 226 92 318 220 102 322 199 120 319
  2 90 50 140 81 60 141 77 62 139
  3 69 24 93 64 29 93 69 26 95
 ER/HER2 subtype
  ER-HER2- 133 72 205 < 0.001 114 89 203 < 0.001 110 100 210 0.001
  ER−HER2+ 74 49 123 79 52 131 70 61 131
  ER+HER2− 547 157 704 509 206 715 445 246 691
  ER+HER2+ 54 31 85 56 32 88 45 42 87
 Age
  Mean 55.2 53.2 54.6 0.015 540.8 540.4 540.6 0.634 540.4 540.4 540.4 0.955
  SD 12.8 12.8 12.8 120.6 130.0 120.8 120.7 120.9 120.8
  Median 53 51 52 52 53 53 52 53 52
  Range 27–97 22–97 22–97 23–94 27–97 22–94
 Tumor size
  Mean 2.7 2.8 2.7 < 0.001 20.7 20.8 20.7 < 0.001 20.7 20.7 20.7 0.610
  SD 1.57 1.25 1.49 10.58 10.3 10.50 10.60 10.27 10.48
  Median 2.3 2.5 2.5 20.2 20.5 20.4 20.4 20.5 20.4
  Range 0.1–13.0 1.0–8.0 00.1–130.0 10.0–80.0 00.1–130.0 00.1–80.0
Biomarkers
 ER
  Negative 207 122 329 < 0.001 195 141 336 < 0.001 181 161 342 0.002
  Positive 601 188 789 565 238 803 490 288 778
 PR
  Negative 253 128 381 0.002 235 148 383 0.007 207 177 384 0.003
  Positive 553 181 734 521 231 752 462 271 733
 Ki67
  Low 634 193 827 < 0.001 580 256 836 0.001 519 297 816 < 0.001
  High 169 117 286 173 121 294 147 151 298
 HER2
  Negative 680 229 909 < 0.001 623 295 918 0.079 558 346 904 0.015
  Positive 128 80 208 135 84 219 115 103 218
 EGFR
  Negative 769 277 1046 < 0.001 710 353 1063 0.590 638 406 1044 0.006
  Positive 30 31 61 40 23 63 29 37 66
 C-kit
  Negative 692 248 940 0.011 635 319 954 0.789 564 373 937 0.456
  Positive 107 60 167 114 60 174 101 75 176
 P63
  Negative 786 286 1072 < 0.001 731 358 1089 0.178 650 422 1072 0.021
  Positive 18 22 40 24 18 42 19 25 44
 CK5/6
  Negative 714 250 964 < 0.001 676 307 983 < 0.001 591 374 965 0.011
  Positive 87 59 146 76 70 146 76 75 151
 CK14
  Negative 763 281 1044 0.014 718 347 1065 0.016 638 412 1050 0.048
  Positive 39 27 66 35 31 66 32 34 66
 AR
  Negative 380 168 548 0.099 349 205 554 0.036 317 235 552 0.186
  Positive 369 130 499 351 157 508 306 192 498
 Vimentin
  Negative 665 236 901 < 0.001 626 294 920 < 0.001 545 357 902 0.022
  Positive 81 61 142 75 68 143 73 72 145
 P-cadherin
  Negative 602 199 801 < 0.001 571 245 816 < 0.001 491 311 802 0.008
  Positive 142 98 240 122 117 239 124 116 240
 pAKT
  Low 542 197 739 0.037 512 233 745 0.004 449 282 731 0.005
  High 182 91 273 166 114 280 143 134 277
 HLA-B
  Low 647 86 733 < 0.001
  High 144 223 367
 HLA-C
  Low 593 48 641 < 0.001 539 111 650 < 0.001
  High 169 257 426 179 255 434

Bold values indicate that p < 0.05

Italic values indicate that p < 0.01

The relationship between combined expression of the HLAs and clinicopathologic features and biomarkers was quite similar to the single HLA. All high HLAs was associated with higher grade (p < 0.001), absence of EIC (p = 0.007), the presence of necrosis (p < 0.001), higher TIL (p < 0.001), higher pT stage (p = 0.003), and differential expression in ER/HER2 subtypes (p < 0.001). Higher level was found in the ER-HER2- (27.4%) and ER-HER2+ (23.1%) than that in ER + HER2- (14.1%) and ER+ HER2+ (17.5%) cases (Table 2). For biomarkers, there were negative association with ER (p < 0.001), PR (p = 0.001) and AR (p = 0.005) but positive association with Ki67 (p < 0.001), p63 (p = 0.017), CK5/6 (p < 0.001), CK14 (p = 0.011), vimentin (p = 0.002), p-cadherin (p < 0.001) and pAKT (p = 0.019). Comparing between those cases with mixed low/high HLA expression (one to two HLA expression) and those with all high (three HLAs) expression, the associations with higher grade (p < 0.001), absence of EIC (p = 0.009), higher TIL (p = 0.001), higher pT stage (p = 0.017), lower ER (p = 0.001), lower PR (p = 0.006), higher CK5/6 (p < 0.001), lower AR (p = 0.001) and higher p-cadherin (p < 0.001) remained (Table 2). Those cases with all high HLA showed significant association with higher pN stage (p = 0.021). There was no association with the presence of necrosis, Ki67, p63, CK14, vimentin and pAKT.

Table 2.

Correlation of HLA-I co-expression status with clinico-pathological factor

All Low Mixed low/high All high Total p value
All Het vs high
Clinico-pathological features
 Grade
  1 87 48 6 141 < 0.001 < 0.001
  2 225 135 57 418
  3 197 173 125 495
 LVI
  Absent 345 245 129 719 0.979 0.849
  Present 137 95 52 284
 Apocrine
  Absent 482 316 171 959 0.170 0.535
  Present 36 38 17 91
 FF
  Absent 357 275 142 774 0.059 0.448
  Present 141 74 45 260
 EIC
  Absent 397 283 168 848 0.007 0.009
  Present 106 68 20 194
 Necrosis
  Absent 409 266 132 807 < 0.001 0.120
  Present 77 79 54 210
 LI
  Low 350 206 85 641 < 0.001 0.001
  High 41 67 56 164
 pT
  1 226 136 53 415 0.003 0.017
  2 239 193 120 552
  3 32 12 11 55
  4 9 8 5 22
 pN
  0 238 185 80 503 0.957 0.021
  1 149 95 61 305
  2 59 40 33 132
  3 51 22 14 87
 ER/HER2 subtype
  ER−HER2− 79 64 54 197 < 0.001 0.009
  ER−HER2+ 53 40 28 121
  ER+HER2− 345 217 92 654
  ER+HER2+ 32 34 14 80
 Age
  Mean 54.8 54.4 54.3 54.6 0.840 0.909
  SD 12.8 12.8 12.9 12.8
  Median 52 52 53 52
  Range 27–97 22–91 23–94
 Tumor size
  Mean 2.74 2.61 2.90 2.73 0.006 0.004
  SD 1.66 1.26 1.29 1.47
  Median 2.3 2.4 2.5 2.5
  Range 0.1–13.0 0.1–8.0 0.1–8.0
Biomarkers
 ER
  Negative 132 105 82 319 < 0.001 0.001
  Positive 377 251 106 734
 PR
  Negative 156 120 86 362 0.001 0.006
  Positive 352 235 102 689
 Ki67
  Low 405 241 126 772 < 0.001 0.937
  High 101 114 62 277
 HER2
  Negative 424 281 146 851 0.144 0.686
  Positive 85 74 42 201
 EGFR
  Negative 484 328 171 983 0.064 0.393
  Positive 21 23 16 60
 C-kit
  Negative 435 295 156 886 0.420 0.916
  Positive 70 59 32 161
 P63
  Negative 496 340 174 1010 0.017 0.128
  Positive 12 14 13 39
 CK5/6
  Negative 454 311 141 906 < 0.001 < 0.001
  Positive 51 44 47 142
 CK14
  Negative 486 332 168 986 0.011 0.128
  Positive 21 23 19 63
 AR
  Negative 240 163 115 518 0.005 0.001
  Positive 227 173 67 467
 Vimentin
  Negative 417 287 144 848 0.002 0.067
  Positive 49 49 38 136
 P-cadherin
  Negative 380 261 115 759 < 0.001 < 0.001
  Positive 85 74 67 226
 pAKT
  Low 351 222 123 696 0.019 0.822
  High 105 102 54 261

Bold values indicate that p < 0.05

Relationship with patient outcome

Follow-Up data were available for 1039 cases. The mean follow-up duration was 67.7 months (range 1–210 months). Among them, 173 cases (16.6%) resulted in breast cancer specific mortality or relapse. Kaplan–meier analysis on each HLA expression did not show significant overall survival differences. However, in HER2+ cancers, HLA-A high expression was associated with longer BCSS (log-rank = 4.553, p = 0.033) while high HLA-C expression was associated with longer BCSS (log-rank = 3.970, p = 0.046) and DFS (log-rank = 5.296, p = 0.021) (Fig. 1a). In addition, all high HLA expression showed the best survival (compared to all low HLA cases: BCSS log-rank = 4.694, p = 0.030; DFS log-rank = 5.153, p = 0.023) (Fig. 1b). Multivariate cox hazard analysis including age, grade, TIL, LVI, pT stage, pN stage, TIL level, HR status, Ki67 and HLA co-expression status demonstrated a trend of HLA co-expression status being a factor for longer DFS (HR = 0.821, p = 0.085, 95% CI = 0.655–1.028) and BCSS (HR = 0.809, p = 0.085, 95% CI = 0.635–1.030) (Table 3). The HLA co-expression could affect the prognostic value of TIL in HER2+ cases. High TIL was associated with significantly better BCSS (log-rank = 4.260, p = 0.036) and marginally DFS (log-rank = 3.760, p = 0.053) (supplementary figure S2). It is interested to note that only those cases with all HLA high expression and high TIL showed significantly better survival (Fig. 1c).

Fig. 1.

Fig. 1

Kaplan–Meier analysis of DFS and BCSS in HER2 + cancers according to (a) individual HLA expression, (b) their co-expression status and (c) their co-expression with TIL level

Table 3.

Cox regression analysis for DFS and BCSS in HER2+ cancers

Feature Hazard ratio Lower 95% CI Upper 95% CI p-value
DFS
 Age 0.971 0.945 0.997 0.028
 Grade 2.294 0.992 5.309 0.052
 pN 2.172 1.622 2.909  < 0.001
 HLA status 0.821 0.655 1.028 0.085
BCSS
 Grade 2.407 0.902 6.422 0.079
 pN 2.173 1.590 2.970  < .001
 HLA status 0.809 0.635 1.030 0.085

Initial factors: age, grade, pT, pN, LVI, TIL20, Ki67 status, luminal subtype, HLA status (All low, mixed low/high, All high)

Bold values indicate that p < 0.05

Expression in nodal metastases and corresponding primary tumors

Among the 314 breast carcinomas and matched nodal metastases, high HLA-A, B and C expression was found in 31.2% (88/282), 35.1% (100/285) and 40.5% (113/279) of primary breast cancers respectively; while only 24.4% (69/282), 25.2% (72/285), and 25.8% (72/279) of the metastases showed high expression of HLA-A, B and C respectively. There was a significant down-regulation in all HLA in nodal metastases (p = 0.020 for HLA-A and p < 0.001 for HLA-B and C) (supplementary table S3). This down-regulation of individual HLA correlated with each other (p < 0.001). Among them, 184 (72.2%) cases showed low HLA expression in all loci at both primary tumor and metastases. Thirty-one cases (12.2%), 29 cases (11.4%) and 21 cases (8.2%) showed concordant high expression at both sites for single, two and all three loci respectively. Interestingly, patients with concordant high expression of all three HLAs in both primary tumors and metastases showed the best survival (DFS compared to none, one and two HLA concordant: log-rank = 6.040, p = 0.014; log-rank = 3.414, p = 0.065; log-rank = 4.139, p = 0.042 respectively) (Fig. 2). In multivariate analysis, concordant primary and nodal metastases expression was demonstrated to be an independently favorable prognostic feature (DFS: HR = 0.741, p < 0.001, 95% CI = 0.597–0.853; BCSS: HR = 0.699, p = 0.003, 95% CI = 0.553–0.883) (Table 4).

Fig. 2.

Fig. 2

Kaplan–Meier analysis of DFS and BCSS in node positive cancer according to the number of HLA loci with concordant high expression in both primary tumor and nodal metastasis

Table 4.

Cox-regression analysis for DFS and BCSS in node positive cases

Feature Hazard ratio Lower 95% CI Upper 95% CI p value
DFS
 Age 0.968 0.947 0.989 0.003
 Grade 1.837 1.103 3.060 0.019
 pN stage 2.139 1.539 2.973 < .001
 PR status 0.384 0.219 0.673 0.001
 HLA T-LN status 0.714 0.597 0.853 < 0.001
BCSS
 Grade 2.217 0.956 5.142 0.064
 pN 2.673 1.775 4.023 < .001
 HER2 status 2.310 1.143 4.666 0.020
 ER status 0.461 0.217 0.980 0.044
 HLA T-LN status 0.699 0.553 0.883 0.003

Initial factors: age, grade, pT, pN, LVI, Ki67, HER2, ER, PR status, HLA T (primary tumor)-LN (nodal metastasis) status (none, single HLA T-LN concordant hi, two HLA T-LN concordant hi and three HLA T-LN concordant hi)

Bold values indicate that p < 0.05

Discussion

Evasion of immune destruction is one of the important traits that promotes tumor transformation. The recently developed immunotherapies aim to overcome the immune escape and reinvigorate the anti-tumor immune function. However, tumor regression can only be achieved in a proportion of patients, indicating that treatment outcome may be determined by both the immunologic features of the host as well as the immune evasion mechanism of the tumor cells.

One of the fundamental mechanisms by which many cancers, including breast cancer, evade immune surveillance is by down-regulation of their HLA-I expression [4]. Low expression in all three HLA-I loci was observed in the current cohort, with the expression rates of HLA-A, B and C being 28.9%, 33.7% and 39.8% respectively. Expression of individual HLA-I loci was associated with features of high grade breast cancers, including the presence of necrosis, high TIL level, negative hormonal receptor expression, higher Ki67, basal markers and pAKT. Similar association of HLA-I expression with high grade tumor were also reported previously [8, 9]. In line with that, IFNγ signaling is reported to be more robust in breast cancer lacking PR [17]. Although some earlier reports with small cohort or using specific cell model suggested an inverse correlation of HLA-I with HER2 expression [18, 19], in our study as well as some others [5, 710], this was not consistently observed. In addition, there appeared to be a lack of association of HLA-B expression with growth receptor expression. The difference could be due to the additional SP1 regulatory elements in HLA-B locus. HER2 signaling could lead to phosphorylation of SP1 and later its transcriptional activity [20].

All HLA low expression was found in 48.3% of the cases. Only 17.9% showed all high HLA expression and 33.8% showed mixed low/high expression of down-regulation of at least one HLA-I locus. Previous studies mainly used anti-pan HLA antibodies [513]. Our results from co-expression analysis agrees with the previously reported down-regulation rate of 47% or more. However, these earlier studies did not differentiate between tumors with mixed low/high and all high HLA expression. We observed different clinico-pathologic features and, importantly, prognostic implication between cases with different co-expression patterns. In fact, each HLA-I allele was associated with different peptide binding repertoires [21]. Loss/down-regulation of any HLA-I locus would be predicted to present a smaller and less diverse repertoire of tumor-derived antigen to immune cells as compared to those with high expression in all loci; thus with a smaller activated T cell repertoire. In addition, selective loss/ downregulation of HLA loci could result in both downregulation of T cell and NK cell mediated tumor cytotoxicity. HLA-I could be ligands for inhibitory NK cell receptors that signal NK cells to remain silent to self antigen. Total loss of HLA can potentially allow tumor cells to become susceptible to NK cell killing. However, in cases with selective HLA loci downregulation (i.e. mixed low/high phenotype), the tumor cells may escape from both T cell and NK cell mediated cytotoxicity. In line with that, recent data demonstrated a progressive decrease in pCR rate from HLA-negative to HLA-low and HLA-normal HER2+ cases while HLA-high group showed the highest response [22]. Compared to mixed high/low cases, cases with all high expression demonstrated the best survival in the HER2+ subset and were associated with a higher level of TIL. All high expression may represent an intact antigen presenting mechanism; thus enabling an efficient activation of anti-tumor immunity and control of tumor progression. Significance of prognostic value for assessing the co-expression pattern of HLA loci could be suggested.

It is interesting to note that the prognostic value of the HLA co-expression could be affected by the TIL level and HLA expression pattern on nodal metastases. In HER2+ cancers, high TIL predicted benefit of anthracycline chemotherapy, trastuzumab treatment and better prognosis [2325]. Of note, only those cases with high TIL together with homogeneous HLA high expression in our cohort demonstrated good prognosis while those with low TIL or low/ heterogeneous HLA had poor survival. The high TIL level indicated the presence of immune surveillance. However, to elicit the effector functions, antigen presentation by HLA expressed on tumor cells is an important pre-requisite. Therefore, the prognostication of TIL should be evaluated in the context of HLA status of the tumor cells. Combined assessment of TIL and HLA expression may provide more accurate prognostic and predictive information. Moreover, downregulation of HLA expression in nodal metastases compared to primary tumor was observed in this study. Favorable outcome was found with cases showing high expression of HLA at both sites, in particular, cases with all high HLA expression in all three loci. Nodal metastasis is one of the most important indicators of poor prognosis, as it is thought that cancer cells from metastatic lymph nodes can further escape into the circulation by invading nodal blood vessels leading to the development of distant metastases [26]. Downregulation of HLA or antigen presenting machinery on metastatic tumors has long been described in metastatic breast cancers [2729]. The current results fit the concept that HLA loss may favor metastatic colonization. Thus, a better outcome could be observed in tumors maintaining high HLA expression in both primary tumor and nodal metastases. This HLA expression pattern could potentially identify node positive patients with better outcome.

Interaction between tumor cells and the immune microenvironment is of pivotal importance in inducing an effective anti-tumor immune response. The loss of HLA-I expression on cancer cells could be a hurdle for successful immunotherapy. It has become increasingly evident that HLA-I phenotype could determine the clinical success of immune checkpoint blockade. Mutations in IFNγ signaling pathway and β2-microglobulin which are required for HLA expression or upregulation were associated with acquired resistance to PD-1 blockade in melanoma [30]. In a cohort of melanoma patients treated with immune checkpoint blockade (ICB), patients with loss of heterozygosity (LOH) or homozygosity in at least one HLA-I locus had reduced survival, independent of somatic mutational load, tumor stage, age or types of treatment [31]. Success of antitumor response unleashed by ICB could depend on the efficacy of tumor antigen presentation to immune cells. An intact HLA phenotype is required for the presentation of a diverse repertoire. Tumor mutational burden (TMB) has been associated with ICB benefit and is currently considered to be a potential biomarker for ICB [32]. However, a higher TMB in tumors with HLA LOH was observed compared with those without HLA LOH in lung cancer patients enrolled in the TRACERx study [33]. One could speculate that patients with a high TMB and HLA LOH may not respond equally to ICB as those without LOH. All these indicated that HLA-I status/expression should be taken into account to predict treatment response. In term of therapy, strategies to restore HLA-I expression for efficient antigen presentation can be helpful. The immunogenic modulation on tumor immune environment, such as up-regulation of inflammatory cytokines and Fas expression as well as an increased MHC I molecules, by low-dose chemotherapy and radiation have been suggested [34, 35]. In a recent clinical trial for metastatic TNBC, induction treatment with short-term chemotherapy or irradiation followed by ICB led to clinical benefit in a substantial subset of patients, with higher than expected response rate and durable responses [36]. Further studies exploring the relationship of HLA and ICB are warrant to provide scope for improved treatment strategies and prediction of treatment responses.

Limitations of the study included the choice of locus specific antibodies. Although the antibodies were raised against specific locus and their application has been verified in other studies, it is possible that they may recognize some specific alleles in the same locus with the lower efficiency due to variability of the regions. Further characterization of the antibodies with a full panel of HLA-I alleles will be required to examine the antibody sensitivity. Nonetheless, the study provide some initial data deserve further validation for its potential clinical utility.

In summary, a combined analysis on the co-expression status of HLA-I loci may be important for the prognostication of HLA expression. It can be incorporated into TIL evaluation and nodal status to provide improved prognostication. In addition, a better outcome could be observed in tumors maintaining high HLA expression in both primary tumor and nodal metastases highlighting the importance of its evaluation on both primary and metastatic tumors. In the era of immunotherapy, HLA-I could to be an important component to be considered. Strategies to promote and assess its expression may be essential.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Acknowledgements

We thanked the Core Utilities for Cancer Genomics and Pathobiology, Anatomical and Cellular Pathology, The Chinese University of Hong Kong for their excellent technical support.

Author contributions

JT analyzed the data and wrote the paper; CH, YS, IP, MM performed the experiments; S-KC, S-YC, KS collected and arranged clinicopathological data of cases; GT conceived the idea for the paper, provided guidance and critically revised the paper. All authors read and approved the final version to be published.

Compliance of ethical standards

Conflicts of interest

All author declared no conflict of interest.

Human and animal right statement

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. The study was approved by Joint Chinese University of Hong Kong- New Territories East Cluster clinical research ethics committee (Re: 2014.500).

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Ghahremanloo A, Soltani A, Modaresi SMS, Hashemy SI. Recent advances in the clinical development of immune checkpoint blockade therapy. Cell Oncol (Dordr) 2019 doi: 10.1007/s13402-019-00456-w. [DOI] [PubMed] [Google Scholar]
  • 2.Atezolizumab Combo Approved for PD-L1-positive TNBC (2019). Cancer Discov 9 (5):OF2. 10.1158/2159–8290.CD-NB2019–038 [DOI] [PubMed]
  • 3.Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora AM, van Hall T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr Opin Immunol. 2016;39:44–51. doi: 10.1016/j.coi.2015.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Garrido F, Cabrera T, Aptsiauri N. "Hard" and "soft" lesions underlying the HLA class I alterations in cancer cells: implications for immunotherapy. Int J Cancer. 2010;127(2):249–256. doi: 10.1002/ijc.25270. [DOI] [PubMed] [Google Scholar]
  • 5.de Kruijf EM, van Nes JG, Sajet A, Tummers QR, Putter H, Osanto S, Speetjens FM, Smit VT, Liefers GJ, van de Velde CJ, Kuppen PJ. The predictive value of HLA class I tumor cell expression and presence of intratumoral Tregs for chemotherapy in patients with early breast cancer. Clin Cancer Res. 2010;16(4):1272–1280. doi: 10.1158/1078-0432.CCR-09-1844. [DOI] [PubMed] [Google Scholar]
  • 6.Gudmundsdottir I, Gunnlaugur Jonasson J, Sigurdsson H, Olafsdottir K, Tryggvadottir L, Ogmundsdottir HM. Altered expression of HLA class I antigens in breast cancer: association with prognosis. Int J Cancer. 2000;89(6):500–505. doi: 10.1002/1097-0215(20001120)89:6&#x0003c;500::AID-IJC6&#x0003e;3.0.CO;2-#. [DOI] [PubMed] [Google Scholar]
  • 7.Kaneko K, Ishigami S, Kijima Y, Funasako Y, Hirata M, Okumura H, Shinchi H, Koriyama C, Ueno S, Yoshinaka H, Natsugoe S. Clinical implication of HLA class I expression in breast cancer. BMC Cancer. 2011;11:454. doi: 10.1186/1471-2407-11-454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Park HS, Cho U, Im SY, Yoo CY, Jung JH, Suh YJ, Choi HJ. Loss of human leukocyte antigen class I expression is associated with poor prognosis in patients with advanced breast cancer. J Pathol Transl Med. 2019;53(2):75–85. doi: 10.4132/jptm.2018.10.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lee HJ, Song IH, Park IA, Heo SH, Kim YA, Ahn JH, Gong G. Differential expression of major histocompatibility complex class I in subtypes of breast cancer is associated with estrogen receptor and interferon signaling. Oncotarget. 2016;7(21):30119–30132. doi: 10.18632/oncotarget.8798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Garrido MA, Rodriguez T, Zinchenko S, Maleno I, Ruiz-Cabello F, Concha A, Olea N, Garrido F, Aptsiauri N. HLA class I alterations in breast carcinoma are associated with a high frequency of the loss of heterozygosity at chromosomes 6 and 15. Immunogenetics. 2018;70(10):647–659. doi: 10.1007/s00251-018-1074-2. [DOI] [PubMed] [Google Scholar]
  • 11.Madjd Z, Spendlove I, Pinder SE, Ellis IO, Durrant LG. Total loss of MHC class I is an independent indicator of good prognosis in breast cancer. Int J Cancer. 2005;117(2):248–255. doi: 10.1002/ijc.21163. [DOI] [PubMed] [Google Scholar]
  • 12.Torigoe T, Asanuma H, Nakazawa E, Tamura Y, Hirohashi Y, Yamamoto E, Kanaseki T, Hasegawa T, Sato N. Establishment of a monoclonal anti-pan HLA class I antibody suitable for immunostaining of formalin-fixed tissue: unusually high frequency of down-regulation in breast cancer tissues. Pathol Int. 2012;62(5):303–308. doi: 10.1111/j.1440-1827.2012.02789.x. [DOI] [PubMed] [Google Scholar]
  • 13.Vertuani S, Triulzi C, Roos AK, Charo J, Norell H, Lemonnier F, Pisa P, Seliger B, Kiessling R. HER-2/neu mediated down-regulation of MHC class I antigen processing prevents CTL-mediated tumor recognition upon DNA vaccination in HLA-A2 transgenic mice. Cancer Immunol Immunother. 2009;58(5):653–664. doi: 10.1007/s00262-008-0587-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Benevolo M, Mottolese M, Piperno G, Sperduti I, Cione A, Sibilio L, Martayan A, Donnorso RP, Cosimelli M, Giacomini P. HLA-A, -B, -C expression in colon carcinoma mimics that of the normal colonic mucosa and is prognostically relevant. Am J Surg Pathol. 2007;31(1):76–84. doi: 10.1097/01.pas.0000213343.55605.b9. [DOI] [PubMed] [Google Scholar]
  • 15.de Jong RA, Boerma A, Boezen HM, Mourits MJ, Hollema H, Nijman HW. Loss of HLA class I and mismatch repair protein expression in sporadic endometrioid endometrial carcinomas. Int J Cancer. 2012;131(8):1828–1836. doi: 10.1002/ijc.27449. [DOI] [PubMed] [Google Scholar]
  • 16.Tsang JY, Au WL, Lo KY, Ni YB, Hlaing T, Hu J, Chan SK, Chan KF, Cheung SY, Tse GM. PD-L1 expression and tumor infiltrating PD-1+ lymphocytes associated with outcome in HER2+ breast cancer patients. Breast Cancer Res Treat. 2017;162(1):19–30. doi: 10.1007/s10549-016-4095-2. [DOI] [PubMed] [Google Scholar]
  • 17.Goodman ML, Trinca GM, Walter KR, Papachristou EK, D'Santos CS, Li T, Liu Q, Lai Z, Chalise P, Madan R, Fan F, Markiewicz MA, Jin VX, Carroll JS, Hagan CR. Progesterone receptor attenuates STAT1-mediated IFN signaling in breast cancer. J Immunol. 2019;202(10):3076–3086. doi: 10.4049/jimmunol.1801152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Choudhury A, Charo J, Parapuram SK, Hunt RC, Hunt DM, Seliger B, Kiessling R. Small interfering RNA (siRNA) inhibits the expression of the Her2/neu gene, upregulates HLA class I and induces apoptosis of Her2/neu positive tumor cell lines. Int J Cancer. 2004;108(1):71–77. doi: 10.1002/ijc.11497. [DOI] [PubMed] [Google Scholar]
  • 19.Inoue M, Mimura K, Izawa S, Shiraishi K, Inoue A, Shiba S, Watanabe M, Maruyama T, Kawaguchi Y, Inoue S, Kawasaki T, Choudhury A, Katoh R, Fujii H, Kiessling R, Kono K. Expression of MHC Class I on breast cancer cells correlates inversely with HER2 expression. Oncoimmunology. 2012;1(7):1104–1110. doi: 10.4161/onci.21056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Li G, Xie Q, Yang Z, Wang L, Zhang X, Zuo B, Zhang S, Yang A, Jia L. Sp1-mediated epigenetic dysregulation dictates HDAC inhibitor susceptibility of HER2-overexpressing breast cancer. Int J Cancer. 2019;145(12):3285–3298. doi: 10.1002/ijc.32425. [DOI] [PubMed] [Google Scholar]
  • 21.Paul S, Weiskopf D, Angelo MA, Sidney J, Peters B, Sette A. HLA class I alleles are associated with peptide-binding repertoires of different size, affinity, and immunogenicity. J Immunol. 2013;191(12):5831–5839. doi: 10.4049/jimmunol.1302101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Muntasell A, Rojo F, Servitja S, Rubio-Perez C, Cabo M, Tamborero D, Costa-Garcia M, Martinez-Garcia M, Menendez S, Vazquez I, Lluch A, Gonzalez-Perez A, Rovira A, Lopez-Botet M, Albanell J. NK cell infiltrates and HLA class I expression in primary HER2(+) breast cancer predict and uncouple pathological response and disease-free survival. Clin Cancer Res. 2019;25(5):1535–1545. doi: 10.1158/1078-0432.CCR-18-2365. [DOI] [PubMed] [Google Scholar]
  • 23.Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, Rouas G, Francis P, Crown JP, Hitre E, de Azambuja E, Quinaux E, Di Leo A, Michiels S, Piccart MJ, Sotiriou C. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02–98. J Clin Oncol. 2013;31(7):860–867. doi: 10.1200/JCO.2011.41.0902. [DOI] [PubMed] [Google Scholar]
  • 24.Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, Kellokumpu-Lehtinen PL, Bono P, Kataja V, Desmedt C, Piccart MJ, Loibl S, Denkert C, Smyth MJ, Joensuu H, Sotiriou C. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol. 2014;25(8):1544–1550. doi: 10.1093/annonc/mdu112. [DOI] [PubMed] [Google Scholar]
  • 25.Perez EA, Ballman KV, Tenner KS, Thompson EA, Badve SS, Bailey H, Baehner FL. Association of stromal tumor-infiltrating lymphocytes with recurrence-free survival in the N9831 adjuvant trial in patients with early-stage HER2-positive breast cancer. JAMA Oncol. 2016;2(1):56–64. doi: 10.1001/jamaoncol.2015.3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pereira ER, Kedrin D, Seano G, Gautier O, Meijer EFJ, Jones D, Chin SM, Kitahara S, Bouta EM, Chang J, Beech E, Jeong HS, Carroll MC, Taghian AG, Padera TP. Lymph node metastases can invade local blood vessels, exit the node, and colonize distant organs in mice. Science. 2018;359(6382):1403–1407. doi: 10.1126/science.aal3622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cordon-Cardo C, Fuks Z, Drobnjak M, Moreno C, Eisenbach L, Feldman M. Expression of HLA-A, B, C antigens on primary and metastatic tumor cell populations of human carcinomas. Cancer Res. 1991;51(23 Pt 1):6372–6380. [PubMed] [Google Scholar]
  • 28.Vitale M, Rezzani R, Rodella L, Zauli G, Grigolato P, Cadei M, Hicklin DJ, Ferrone S. HLA class I antigen and transporter associated with antigen processing (TAP1 and TAP2) down-regulation in high-grade primary breast carcinoma lesions. Cancer Res. 1998;58(4):737–742. [PubMed] [Google Scholar]
  • 29.Liu Y, Komohara Y, Domenick N, Ohno M, Ikeura M, Hamilton RL, Horbinski C, Wang X, Ferrone S, Okada H. Expression of antigen processing and presenting molecules in brain metastasis of breast cancer. Cancer Immunol Immunother. 2012;61(6):789–801. doi: 10.1007/s00262-011-1137-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly L, Saco J, Homet Moreno B, Mezzadra R, Chmielowski B, Ruchalski K, Shintaku IP, Sanchez PJ, Puig-Saus C, Cherry G, Seja E, Kong X, Pang J, Berent-Maoz B, Comin-Anduix B, Graeber TG, Tumeh PC, Schumacher TN, Lo RS, Ribas A. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):819–829. doi: 10.1056/NEJMoa1604958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, Kuo F, Kendall SM, Requena D, Riaz N, Greenbaum B, Carroll J, Garon E, Hyman DM, Zehir A, Solit D, Berger M, Zhou R, Rizvi NA, Chan TA. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359(6375):582–587. doi: 10.1126/science.aao4572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, Peters S. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30(1):44–56. doi: 10.1093/annonc/mdy495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins TBK, Wilson GA, Birkbak NJ, Veeriah S, Van Loo P, Herrero J, Swanton C, Consortium TR (2017) Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell 171 (6):1259–1271e1211. doi:10.1016/j.cell.2017.10.001. [DOI] [PMC free article] [PubMed]
  • 34.Kershaw MH, Devaud C, John LB, Westwood JA, Darcy PK. Enhancing immunotherapy using chemotherapy and radiation to modify the tumor microenvironment. Oncoimmunology. 2013;2(9):e25962. doi: 10.4161/onci.25962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kwilas AR, Donahue RN, Bernstein MB, Hodge JW. In the field: exploiting the untapped potential of immunogenic modulation by radiation in combination with immunotherapy for the treatment of cancer. Front Oncol. 2012;2:104. doi: 10.3389/fonc.2012.00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de Vijver KK, de Maaker M, Nederlof I, Kluin RJC, Warren S, Ong S, Wiersma TG, Russell NS, Lalezari F, Schouten PC, Bakker NAM, Ketelaars SLC, Peters D, Lange CAH, van Werkhoven E, van Tinteren H, Mandjes IAM, Kemper I, Onderwater S, Chalabi M, Wilgenhof S, Haanen J, Salgado R, de Visser KE, Sonke GS, Wessels LFA, Linn SC, Schumacher TN, Blank CU, Kok M. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med. 2019;25(6):920–928. doi: 10.1038/s41591-019-0432-4. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES