The relatively low success rate of phase II oncology trials in predicting success of novel drugs in phase III trials and in gaining regulatory approval may be due to reliance on the endpoint of response rate defined by the RECIST. The neoadjuvant treatment paradigm allows the antitumor activity of a novel therapy to be determined on a pathologic basis at the time of surgery instead of by RECIST, which was not developed to guide clinical decision making or correlate with long-term outcomes. Indeed, the FDA endorsed pathologic complete response (pCR) as a surrogate for overall survival (OS) in early-stage breast cancer and granted accelerated approval to pertuzumab based on this endpoint. We propose that pCR is a biologically rational method of determining treatment effect that may be more likely to predict OS. We discuss some advantages of the neoadjuvant trial design, review the use of neoadjuvant therapy as standards of care, and consider the neoadjuvant platform as a method for drug development. Clin Cancer Res; 22(10); 2323–8. ©2016 AACR.

The primary objective of phase II trials in oncology has been to determine whether a novel drug has sufficient antitumor activity to warrant further investigation in phase III trials. Because of the cost and number of patients required, the failure of a phase III trial has detrimental financial and human ramifications. Most observers have concluded that phase II studies in oncology have had a poor track record in predicting success in phase III trials as well as eventual regulatory approval (1). By one estimate, only 57% of oncology drugs taken from phases II to III obtain FDA approval, a proportion notably lower than for non-oncology drugs (2). The likelihood that a positive phase II trial of combination therapy will result in a subsequent trial that improves the standard of care within 5 years has been reported to be only 0.038 (3).

This low predictive value of phase II trials may be due to reliance on the endpoint of response rate (RR) as defined by the RECIST (2). RECIST was based on work done decades ago to standardize tumor responses in the absence of modern cross-sectional imaging techniques (4, 5). Criteria for partial responses (PR) were based on the precision with which oncologists could differentiate solid spheres of different sizes under a layer of foam rubber (4), not based on clinical outcomes. There was never any claim that the PR criteria were correlated with clinically meaningful outcomes. In fact, the original WHO response criteria (5) and later RECIST (6) stressed that “it is not intended that these RECIST guidelines play a role in…decision making, except if determined appropriate by the treating oncologist” (6). Therefore, it is not surprising that RR has not been a reliable endpoint for phase II trials (7–9).

It seems reasonable to speculate that pathologic complete response (pCR) might correlate more strongly with overall survival (OS) than RRs defined by RECIST. pCR is a biologically rational reflection of a therapy's ability to eradicate metastatic disease, and may therefore serve as a surrogate for OS. Because the number of tumor cells in undetectable micrometastases is many logs lower than the cell number in clinically evident tumors, even pathologic responses less than complete eradication of the detectable tumor might correlate with improved OS. The strategy of neoadjuvant therapy (treatment before complete surgical resection) permits investigators to assess tumor response on a pathologic basis. In this setting, it may be possible to assess treatment benefit in a more meaningful way than in traditional phase II trials using RECIST. In this review, we will consider the use of neoadjuvant trials as a way to identify treatments perhaps more likely to improve OS and as a strategy for drug development.

Different measurements of pathologic response after neoadjuvant therapy have been demonstrated to correlate with OS across various solid tumors (Table 1). The most extensive experience using neoadjuvant therapy is in breast cancer. A strong association between pCR and survival has been demonstrated in many multi-institutional, randomized neoadjuvant trials of chemotherapy in early-stage breast cancer (10–15). However, the definition of pCR varied across these neoadjuvant breast cancer studies and the relationship between pCR and long-term benefit was not always clear. To address these challenges, the FDA performed a pooled analysis of nearly 13,000 patients enrolled in neoadjuvant breast cancer trials (16). The eradication of invasive cancer in both the resected breast tissue and regional lymph nodes was found to correlate more strongly with improved long-term outcomes than was tumor eradication in the breast alone. In addition, the pooled analysis found individual patients who attain a pCR had a 64% reduction in the risk of death compared with patients who did not. At the individual trial level, only a weak association was evident between increases in the proportion of patients achieving a pCR and the ability of treatment to improve OS. However, the heterogeneous patient populations, the low overall rates of pCR, and the lack of targeted therapy in the trials included in the analysis can explain this finding (17). Although the FDA ultimately concluded that pCR meets the surrogate endpoint criterion of being “reasonably likely to predict clinical benefit” (18), future pooled analyses of targeted therapy trials in biomarker-defined breast cancer subtypes could help solidify pCR as a surrogate endpoint for long-term outcomes (19).

Table 1.

Examples of pathologic response measurement that correlate with OS

DiseaseTherapyNPathologic response definitionHR for OSReference
Breast Pooled analysis 2,761 hormone receptor–positive HER2–positive pCR HR, 0.49 (0.33–0.71) (16) 
  1,743 HER2-positive pCR HR, 0.39 (0.31–0.50)  
  1,157 hormone receptor–negative HER2-positive pCR HR, 0.24 (0.18–0.33)  
Breast AC 751 pCR HR, 0.32 P < 0.001 (14) 
 AC and AC-T 2,344 pCR HR, 0.36 P < 0.0001  
Breast Chemotherapy + trastuzumab 217 HER2+ pCR HR 4.9 (1.4–17.4) if no pCR P = 0.012 (54) 
Bladder MVAC 147 <pT2 HR not reported; 5-year landmark OS 75% vs. 30% (21) 
Bladder GC 154 <pT2 HR 6.7 (2.6–17.4) if ≥ pT2 P < 0.001 (23) 
Lung Docetaxel–cisplatin 75 pN0-N1 HR, 0.22 (0.10–0.49) P = 0.0003 (55) 
Lung Variable chemotherapy 192 ≤10% viable tumor HR, 2.39 (0.99–5.78) if > 10% P = 0.05 (26, 27) 
Esophageal and esophagogastric Variable chemotherapy 400 Tumor downstaging HR, 0.43 (0.31–0.59) P = < 0.001 (31) 
DiseaseTherapyNPathologic response definitionHR for OSReference
Breast Pooled analysis 2,761 hormone receptor–positive HER2–positive pCR HR, 0.49 (0.33–0.71) (16) 
  1,743 HER2-positive pCR HR, 0.39 (0.31–0.50)  
  1,157 hormone receptor–negative HER2-positive pCR HR, 0.24 (0.18–0.33)  
Breast AC 751 pCR HR, 0.32 P < 0.001 (14) 
 AC and AC-T 2,344 pCR HR, 0.36 P < 0.0001  
Breast Chemotherapy + trastuzumab 217 HER2+ pCR HR 4.9 (1.4–17.4) if no pCR P = 0.012 (54) 
Bladder MVAC 147 <pT2 HR not reported; 5-year landmark OS 75% vs. 30% (21) 
Bladder GC 154 <pT2 HR 6.7 (2.6–17.4) if ≥ pT2 P < 0.001 (23) 
Lung Docetaxel–cisplatin 75 pN0-N1 HR, 0.22 (0.10–0.49) P = 0.0003 (55) 
Lung Variable chemotherapy 192 ≤10% viable tumor HR, 2.39 (0.99–5.78) if > 10% P = 0.05 (26, 27) 
Esophageal and esophagogastric Variable chemotherapy 400 Tumor downstaging HR, 0.43 (0.31–0.59) P = < 0.001 (31) 

Abbreviations: AC, Adriamycin and cyclophosphamide; T, paclitaxel; <pT2, non–muscle-invasive bladder cancer; ≥pT2, muscle-invasive bladder cancer.

In neoadjuvant bladder cancer trials, pCR, less than CR, has been associated with improved OS (20). In an analysis of 147 patients by Splinter and colleagues (21), patients with muscle-invasive bladder cancer (MIBC) whose disease was downstaged to no muscle invasion (<pT2) after neoadjuvant chemotherapy experienced a 75% survival rate at 5 years compared with 20% survival for those whose tumors still showed muscle invasion (≥pT2 residual disease). The equivalency of pCR and <pT2 in predicting OS after neoadjuvant chemotherapy for bladder cancer was confirmed in a prospective trial (22) and a retrospective analysis (23). The development of the pCR for bladder cancer highlights the importance of considering the disease biology when defining the endpoint. Neoadjuvant phase II trials in MIBC are a model for rational drug development and can use <pT2 as a surrogate endpoint (24).

In other solid tumors, where eradication or significant downstaging of disease after neoadjuvant therapy is a rare event, investigators have used histopathologic methods to categorize response and correlate them with outcome. In stage IB-IIIA non–small cell lung cancer (NSCLC), the median rate of pCR from 15 trials of neoadjuvant chemotherapy was only 4% (range, 0–16%; ref. 25). Pataer and colleagues (26) developed a technique for assessing response to neoadjuvant chemotherapy based on mean a percentage of residual viable tumor cells taken from sampled tissue. In a comprehensive tissue analysis of 192 patients with resected stage I–IV NSCLC given neoadjuvant chemotherapy, a cutoff value of ≤ 10% viable tumor was associated with improvement in OS (HR, 2.39; P = 0.05 if >10% viable tumor) on multivariate analysis (26, 27). These methods of evaluating pCR (26) were applied prospectively in a trial of 50 patients with stage IB–IIIA NSCLC given neoadjuvant chemotherapy and bevacizumab (28). Of the 22% of patients with ≤10% viable tumor, 100% were alive at 3 years compared with only 49% of those who had >10% residual tumor (P = 0.01); this remained statistically significant after adjustment for stage (P = 0.02). Although validation in larger studies across NSCLC histologies is needed, Hellman and colleagues (25) propose that ≤10% residual tumor in resected lung and lymph node tissue should be regarded as a surrogate of OS in patients with resectable NSCLC given neoadjuvant chemotherapy.

These experiences, and others not discussed here (29–31), indicate that neoadjuvant treatment can lead to improved OS depending on how response is defined. Although some trials used pCR as the endpoint, pathologic responses, less than complete responses, may be associated with improved survival in certain tumor types.

Neoadjuvant administration of systemic therapy has the following potential benefits to patients: (i) tumor cytoreduction leading to improved surgical outcome or, in some cases, less radical surgery; (ii) sooner treatment of systemic metastases without the possible barrier of postoperative complications; (iii) ability to determine, in a short period of time, if the tumor is sensitive to the systemic therapy. Indeed, neoadjuvant therapy is used as a standard of care for some tumors in carefully selected patients (29, 32–34).

Breast cancer

In early-stage breast cancer, multiple studies have shown that neoadjuvant therapy improves OS and may also reduce the extent of local surgery required (10, 35–37). The multidisciplinary approach required and the current controversies have been reviewed recently (32). Neoadjuvant therapy appears to be more beneficial in aggressive subtypes such as triple-negative and HER2-positive tumors that are more chemosensitive and have higher pCR rates (15, 38).

Bladder cancer

Neoadjuvant cisplatin–based chemotherapy followed by radical cystectomy is a standard of care for MIBC. Randomized neoadjuvant trials using MVAC (methotrexate, vinblastine, doxorubicin, and cisplatin; ref. 20) or CMV (cisplatin, methotrexate, and vinblastine; ref. 39) before radical cystectomy have both been shown to improve survival compared with cystectomy alone. Patients receiving neoadjuvant MVAC had superior disease specific survival [HR, 1.66; 95% confidence interval (CI), 1.22–2.45; P = 0.002] and a trend toward superior OS (HR, 1.33; 95% CI, 1.00–1.76) compared with patients who were managed with surgery alone, with OS rates of 57% and 43% at 5 years, respectively (P = 0.06). The neoadjuvant CMV trial reported that neoadjuvant CMV was associated with a 16% relative improvement in survival (P = 0.037) and a 23% relative improvement in metastasis-free survival (P = 0.0001) at 10 years. Given that gemcitabine and cisplatin (GC) is better tolerated and achieves similar survival rates to those from MVAC in the metastatic disease setting (40), it is frequently used as a substitute for MVAC in the neoadjuvant setting and has similar pathologic RRs (41). In a meta-analysis of over 3,000 patients with MIBC, there was a 5-year OS benefit seen with neoadjuvant cisplatin–based chemotherapy followed by radical cystectomy versus radical cystectomy alone (HR, 0.86; 95% CI, 0.77–0.95; P = 0.003; ref. 33).

Non–small cell lung cancer

In patients with resectable NSCLC, neoadjuvant chemotherapy can improve OS. In the 1990s, two small, randomized trials were terminated early on the basis of an interim analysis showing significant improvement in survival for neoadjuvant chemotherapy followed by surgery versus surgery alone (42, 43). In a meta-analysis performed by Song and colleagues (34) of 13 randomized controlled trials patients with stage IB–IIIA NSCLC who received neoadjuvant chemotherapy had improved OS compared with patients who were managed with surgery alone (combined HR, 0.84; 95% CI, 0.77–0.92; P = 0.0001). These results were similar to those from a prior meta-analysis, in which a combined HR of 0.82 (95% CI, 0.69–0.97) was obtained. A more recent phase III trial in patients with stage IB–IIIA NSLCC randomly assigned patients to surgery alone or surgery plus neoadjuvant CG (44). The HR for OS was 0.63 (95% CI, 0.43–0.92; P = 0.02), favoring the neoadjuvant chemotherapy arm. In addition, patients with stage IIB/IIIA disease had a survival benefit of 23.4% at 3 years.

Other tumor types

Randomized trials of neoadjuvant chemotherapy in other tumor types have demonstrated an improvement in OS when compared with surgery alone (29, 30, 45, 46). Chemoradiotherapy is also routinely used in the neoadjuvant treatment of some solid tumors (47–49). These experiences further support the notion that pCR in the neoadjuvant setting can serve as a surrogate for OS.

In May 2012, the FDA published a draft guidance outlining the reasons for its acceptance of pCR as a surrogate in early-stage breast cancer and proposing a framework for its use in clinical trials (50). In September 2013, the FDA granted accelerated approval to pertuzumab for use in combination with trastuzumab and docetaxel as neoadjuvant treatment of patients with HER2-positive, early-stage breast cancer. The approval was based on the NeoSphere trial, which demonstrated an improvement in the pCR rate seen with pertuzumab and trastuzumab plus docetaxel compared with trastuzumab plus docetaxel (45.8% vs. 29.0%; P = 0.0141; ref. 51).

Although an improvement in pCR ultimately led to the accelerated approval of pertuzumab, the other factors that were crucial to the FDA decision reveal important, generalizable considerations for using neoadjuvant therapy as a platform for drug development (Fig. 1). First, patients with HER2-positive breast cancer are at high risk for relapse with standard therapy (15). Second, the safety profile of pertuzumab alone and in combination with standard-of-care chemotherapy and trastuzumab had been reported in nearly 10,000 patients (17). Third, the FDA had conducted the aforementioned pooled analysis of neoadjuvant trials to establish the definition of pCR and to support the relationship between pCR and OS at the patient level (16). Finally, the adjuvant APHINITY confirmatory trial was fully accrued and well under way at the time of accelerated approval, complying with the FDA requirement for a postmarketing clinical trial to verify meaningful clinical benefit (50).

Figure 1.

Considerations for designing neoadjuvant trials for drug development and approval.

Figure 1.

Considerations for designing neoadjuvant trials for drug development and approval.

Close modal

The bar for the first approval of a drug based on pCR was set high, but has provided an impetus to pursue this pathway of expedited drug development in breast cancer and other solid tumors. Indeed, once a pCR assessment has been validated as a surrogate in randomized clinical trials, it can be used as an endpoint in nonrandomized trials as well to screen drugs for antitumor activity based on results from historical controls. For example, two recent, single-arm, phase II trials in MIBC have evaluated dose-dense MVAC (ddMVAC) in the neoadjuvant setting (52, 53). On the basis of the pathologic RRs seen in these trials, ddMVAC has been carried forward to randomized clinical trials in the neoadjuvant setting (NCT02177695 and NCT01812369).

Advances in molecularly targeted therapy and immunotherapy have led to an explosion of new drugs for cancer patients. Despite this progress, the field remains reliant on RECIST, an antiquated method of measuring responses never designed to correlate with clinically meaningful outcomes. We speculate that pCR, as assessed in neoadjuvant trials, might be expected to correlate more strongly with OS. Indeed, pCR has already been endorsed by the FDA as a surrogate for OS in early-stage breast cancer (50). However, patient selection will remain important, and the few patients who are thought to have a brief window of opportunity for curative surgery may not be appropriate for neoadjuvant treatment with experimental therapies. Conversely, if a neoadjuvant therapy has already been shown to improve OS, new drugs may need to be tested as add-ons to the standard therapy.

Another advantage of neoadjuvant trials is the availability of pre- and posttreatment tumor tissue for study. Investigation facilitated by the neoadjuvant paradigm has already contributed to our understanding of how tumor genomics and the immune microenvironment can serve as predictive biomarkers of response in solid tumors (Table 2). In patients who do not respond to treatment, the posttreatment tumor can provide critical information about mechanisms of treatment failure. Tumors can be studied for intrinsic resistance or for the failure of the drug treatment to hit the intended molecular target. For immunotherapy treatments, studies of the tumor environment are likely to be very important. The ability to assess the activity of a drug in vivo and to interrogate posttreatment tissue will aid in the discovery of response and resistance mechanisms that can guide the next generation of clinical trials.

Table 2.

Examples of predictive biomarker discovery facilitated by the neoadjuvant paradigm

DiseaseTherapyPredictive biomarkerReference
Breast Anti-HER2 agents PIK3CA mutations (56) 
Breast Chemotherapy BRCA1 mutations (57) 
Breast Anti-HER2 agents and chemotherapy TILs (58–61) 
Bladder Platinum-based chemotherapy DDR deficiency (62, 63) 
Ovarian Platinum-based chemotherapy TILs (64) 
DiseaseTherapyPredictive biomarkerReference
Breast Anti-HER2 agents PIK3CA mutations (56) 
Breast Chemotherapy BRCA1 mutations (57) 
Breast Anti-HER2 agents and chemotherapy TILs (58–61) 
Bladder Platinum-based chemotherapy DDR deficiency (62, 63) 
Ovarian Platinum-based chemotherapy TILs (64) 

Abbreviations: DDR, DNA damage repair; TIL, tumor infiltrating lymphocytes.

S.A. Funt has ownership interest in Kite Pharma. P.B. Chapman reports receiving commercial research support from Bristol-Myers Squibb, Genentech, GlaxoSmithKline, and Novartis and is a consultant/advisory board member for Bristol-Myers Squibb, Genentech, and GlaxoSmithKline. No other potential conflicts of interest were disclosed.

Conception and design: S.A. Funt, P.B. Chapman

Writing, review, and/or revision of the manuscript: S.A. Funt, P.B. Chapman

P.B. Chapman was supported in part by the John K. Figge Research Fund.

1.
Lara
PN
,
Redman
MW
. 
The hazards of randomized phase II trials
.
Ann Oncol
2012
;
23
:
7
9
.
2.
DiMasi
JA
,
Grabowski
HG
. 
Economics of new oncology drug development
.
J Clin Oncol
2007
;
25
:
209
16
.
3.
Maitland
ML
,
Hudoba
C
,
Snider
KL
,
Ratain
MJ
. 
Analysis of the yield of phase II combination therapy trials in medical oncology
.
Clin Cancer Res
2010
;
16
:
5296
302
.
4.
Moertel
CG
,
Hanley
JA
. 
The effect of measuring error on the results of therapeutic trials in advanced cancer
.
Cancer
1976
;
38
:
388
94
.
5.
Miller
AB
,
Hoogstraten
B
,
Staquet
M
,
Winkler
A
. 
Reporting results of cancer treatment
.
Cancer
1981
;
47
:
207
14
.
6.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
7.
Ratain
MJ
,
Eckhardt
SG
. 
Phase II studies of modern drugs directed against new targets: if you are fazed, too, then resist RECIST
.
J Clin Oncol
2004
;
22
:
4442
5
.
8.
Buyse
M
,
Thirion
P
,
Carlson
RW
,
Burzykowski
T
,
Molenberghs
G
,
Piedbois
P
. 
Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis
.
Lancet
2000
;
356
:
373
8
.
9.
Zabor
EC
,
Heller
G
,
Schwartz
LH
,
Chapman
PB
. 
Correlating surrogate endpoints with overall survival at the individual patient level in BRAFV600E-mutated metastatic melanoma patients treated with vemurafenib
.
Clin Cancer Res
2016
;
22
:
1341
7
.
10.
Fisher
B
,
Bryant
J
,
Wolmark
N
,
Mamounas
E
,
Brown
A
,
Fisher
ER
, et al
Effect of preoperative chemotherapy on the outcome of women with operable breast cancer
.
J Clin Oncol
1998
;
16
:
2672
85
.
11.
Guarneri
V
,
Broglio
K
,
Kau
S-W
,
Cristofanilli
M
,
Buzdar
AU
,
Valero
V
, et al
Prognostic value of pathologic complete response after primary chemotherapy in relation to hormone receptor status and other factors
.
J Clin Oncol
2006
;
24
:
1037
44
.
12.
Kuerer
HM
,
Newman
LA
,
Smith
TL
,
Ames
FC
,
Hunt
KK
,
Dhingra
K
, et al
Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy
.
J Clin Oncol
1999
;
17
:
460
9
.
13.
van der Hage
JA
,
van de Velde
CJH
,
Julien
J-P
,
Tubiana-Hulin
M
,
Vandervelden
C
,
Duchateau
L
, et al
Preoperative chemotherapy in primary operable breast cancer: results from the European Organization for Research and Treatment of Cancer Trial 10902
.
J Clin Oncol
2001
;
19
:
4224
37
.
14.
Rastogi
P
,
Anderson
SJ
,
Bear
HD
,
Geyer
CE
,
Kahlenberg
MS
,
Robidoux
A
, et al
Preoperative chemotherapy: updates of National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27
.
J Clin Oncol
2008
;
26
:
778
85
.
15.
Von Minckwitz
G
,
Untch
M
,
Blohmer
J-U
,
Costa
SD
,
Eidtmann
H
,
Fasching
PA
, et al
Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes
.
J Clin Oncol
2012
;
30
:
1796
804
.
16.
Cortazar
P
,
Zhang
L
,
Untch
M
,
Mehta
K
,
Costantino
JP
,
Wolmark
N
, et al
Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis
.
Lancet
2014
;
384
:
164
72
.
17.
Amiri-Kordestani
L
,
Wedam
S
,
Zhang
L
,
Tang
S
,
Tilley
A
,
Ibrahim
A
, et al
First FDA approval of neoadjuvant therapy for breast cancer: pertuzumab for the treatment of patients with HER2-positive breast cancer
.
Clin Cancer Res
2014
;
20
:
5359
64
.
18.
Pazdur
R
. 
Endpoints for assessing drug activity in clinical trials
.
Oncologist
2008
;
13
:
19
21
.
19.
Amiri-Kordestani
L
,
Beaver
JA
,
Cortazar
P
. 
Neoadjuvant therapy as a platform for drug development: current controversies and regulatory perspectives
.
Oncology
2015
;
29
:
843
4, 846
.
20.
Grossman
HB
,
Natale
RB
,
Tangen
CM
,
Speights
VO
,
Vogelzang
NJ
,
Trump
DL
, et al
Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer
.
N Engl J Med
2003
;
349
:
859
66
.
21.
Splinter
TA
,
Scher
HI
,
Denis
L
,
Bukowski
R
,
Simon
S
,
Klimberg
I
, et al
The prognostic value of the pathological response to combination chemotherapy before cystectomy in patients with invasive bladder cancer. European Organization for Research on Treatment of Cancer–Genitourinary Group
.
J Urol
1992
;
147
:
606
8
.
22.
Schultz
PK
,
Herr
HW
,
Zhang
Z-F
,
Bajorin
DF
,
Seidman
A
,
Sarkis
A
, et al
Neoadjuvant chemotherapy for invasive bladder cancer: prognostic factors for survival of patients treated with M-VAC with 5-year follow-up
.
J Clin Oncol
1994
;
12
:
1394
401
.
23.
Tully
CM
,
Bochner
BH
,
Dalbagni
G
,
Zabor
EC
,
Herr
HW
,
Donat
SM
, et al
. 
Gemcitabine-cisplatin (GC) plus radical cystectomy-pelvic lymph node dissection (RC-PLND) for patients (pts) with muscle-invasive bladder cancer (MIBC): assessing impacts of neoadjuvant chemotherapy (NAC) and the PLND
.
J Clin Oncol
32
, 
2014
(suppl 4; abstr 355).
24.
Balar
AV
,
Milowsky
MI
. 
Neoadjuvant therapy in muscle-invasive bladder cancer: a model for rational accelerated drug development
.
Urol Clin North Am
2015
;
42
:
217
24
.
25.
Hellmann
MD
,
Chaft
JE
,
William
WN
 Jr
,
Rusch
V
,
Pisters
KMW
,
Kalhor
N
, et al
Pathological response after neoadjuvant chemotherapy in resectable non–small cell lung cancers: proposal for the use of major pathological response as a surrogate endpoint
.
Lancet Oncol
2014
;
15
:
e42
50
.
26.
Pataer
A
,
Kalhor
N
,
Correa
AM
,
Raso
MG
,
Erasmus
JJ
,
Kim
ES
, et al
Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy
.
J Thorac Oncol
2012
;
7
:
825
32
.
27.
William
WN
,
Pataer
A
,
Kalhor
N
,
Correa
AM
,
Rice
DC
,
Wistuba
II
, et al
Computed tomography RECIST assessment of histopathologic response and prediction of survival in patients with resectable non–small cell lung cancer after neoadjuvant chemotherapy
.
J Thorac Oncol
2013
;
8
:
222
8
.
28.
Chaft
JE
,
Rusch
V
,
Ginsberg
MS
,
Paik
PK
,
Finley
DJ
,
Kris
MG
, et al
Phase II trial of neoadjuvant bevacizumab plus chemotherapy and adjuvant bevacizumab in patients with resectable non-squamous non–small cell lung cancers
.
J Thorac Oncol
2013
;
8
:
1084
90
.
29.
Vergote
I
,
Tropé
CG
,
Amant
F
,
Kristensen
GB
,
Ehlen
T
,
Johnson
N
, et al
Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer
.
N Engl J Med
2010
;
363
:
943
53
.
30.
Cunningham
D
,
Allum
WH
,
Stenning
SP
,
Thompson
JN
,
Van de Velde
CJH
,
Nicolson
M
, et al
Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer
.
N Engl J Med
2006
;
355
:
11
20
.
31.
Davies
AR
,
Gossage
JA
,
Zylstra
J
,
Mattsson
F
,
Lagergren
J
,
Maisey
N
, et al
Tumor stage after neoadjuvant chemotherapy determines survival after surgery for adenocarcinoma of the esophagus and esophagogastric junction
.
J Clin Oncol
2014
;
32
:
2983
90
.
32.
Santa-Maria
CA
,
Camp
M
,
Cimino-Mathews
A
,
Harvey
S
,
Wright
J
,
Stearns
V
. 
Neoadjuvant therapy for early-stage breast cancer: current practice, controversies, and future directions
.
Oncology
2015
;
29
:
828
38
.
33.
Vale
CL
. 
Neoadjuvant chemotherapy in invasive bladder cancer: update of a systematic review and meta-analysis of individual patient data
.
Eur Urol
2005
;
48
:
202
6
.
34.
Song
W-A
,
Zhou
N-K
,
Wang
W
,
Chu
X-Y
,
Liang
C-Y
,
Tian
X-D
, et al
Survival benefit of neoadjuvant chemotherapy in non–small cell lung cancer: an updated meta-analysis of 13 randomized control trials
.
J Thorac Oncol
2010
;
5
:
510
6
.
35.
Group EBCTC. 
Polychemotherapy for early breast cancer: an overview of the randomised trials
.
Lancet
1998
;
352
:
930
42
.
36.
Mauri
D
,
Pavlidis
N
,
Ioannidis
JPA
. 
Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis
.
J Natl Cancer Inst
2005
;
97
:
188
94
.
37.
Fisher
B
,
Anderson
S
,
Bryant
J
,
Margolese
RG
,
Deutsch
M
,
Fisher
ER
, et al
Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer
.
N Engl J Med
2002
;
347
:
1233
41
.
38.
Carey
LA
,
Dees
EC
,
Sawyer
L
,
Gatti
L
,
Moore
DT
,
Collichio
F
, et al
The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes
.
Clin Cancer Res
2007
;
13
:
2329
34
.
39.
International Collaboration of Trialists; Medical Research Council Advanced Bladder Cancer Working Party (now the National Cancer Research Institute Bladder Cancer Clinical Studies Group); European Organisation for Research and Treatment of Cancer Genito-Urinary Tract Cancer Group; Australian Bladder Cancer Study Group; National Cancer Institute of Canada Clinical Trials Group; Finnbladder; et al. 
International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: long-term results of the BA06 30894 trial
.
J Clin Oncol
2011
;
29
:
2171
7
.
40.
von der Maase
H
,
Hansen
SW
,
Roberts
JT
,
Dogliotti
L
,
Oliver
T
,
Moore
MJ
, et al
Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study
.
J Clin Oncol
2000
;
18
:
3068
77
.
41.
Dash
A
,
Pettus
JA
,
Herr
HW
,
Bochner
BH
,
Dalbagni
G
,
Donat
SM
, et al
A role for neoadjuvant gemcitabine plus cisplatin in muscle-invasive urothelial carcinoma of the bladder
.
Cancer
2008
;
113
:
2471
7
.
42.
Rosell
R
,
Gómez-Codina
J
,
Camps
C
,
Javier Sánchez
J
,
Maestre
J
,
Padilla
J
, et al
Preresectional chemotherapy in stage IIIA non-small-cell lung cancer: a 7-year assessment of a randomized controlled trial
.
Lung Cancer
1999
;
26
:
7
14
.
43.
Roth
JA
,
Fossella
F
,
Komaki
R
,
Ryan
MB
,
Putnam
JB
,
Lee
JS
, et al
A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer
.
J Natl Cancer Inst
1994
;
86
:
673
80
.
44.
Scagliotti
GV
,
Pastorino
U
,
Vansteenkiste
JF
,
Spaggiari
L
,
Facciolo
F
,
Orlowski
TM
, et al
Randomized phase III study of surgery alone or surgery plus preoperative cisplatin and gemcitabine in stages IB to IIIA non–small cell lung cancer
.
J Clin Oncol
2012
;
30
:
172
8
.
45.
Ychou
M
,
Boige
V
,
Pignon
J-P
,
Conroy
T
,
Bouché
O
,
Lebreton
G
, et al
Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial
.
J Clin Oncol
2011
;
29
:
1715
21
.
46.
Sjoquist
KM
,
Burmeister
BH
,
Smithers
BM
,
Zalcberg
JR
,
Simes
RJ
,
Barbour
A
, et al
Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis
.
Lancet Oncol
2011
;
12
:
681
92
.
47.
Van Hagen
P
,
Hulshof
MCCM
,
van Lanschot
JJB
,
Steyerberg
EW
,
Henegouwen MI van
B
,
Wijnhoven
BPL
, et al
Preoperative chemoradiotherapy for esophageal or junctional cancer
.
N Engl J Med
2012
;
366
:
2074
84
.
48.
Sauer
R
,
Becker
H
,
Hohenberger
W
,
Rödel
C
,
Wittekind
C
,
Fietkau
R
, et al
Preoperative versus postoperative chemoradiotherapy for rectal cancer
.
N Engl J Med
2004
;
351
:
1731
40
.
49.
Haddad
R
,
O'Neill
A
,
Rabinowits
G
,
Tishler
R
,
Khuri
F
,
Adkins
D
, et al
Induction chemotherapy followed by concurrent chemoradiotherapy (sequential chemoradiotherapy) versus concurrent chemoradiotherapy alone in locally advanced head and neck cancer (PARADIGM): a randomised phase 3 trial
.
Lancet Oncol
2013
;
14
:
257
64
.
50.
Guidance for industry
. 
Pathological complete response in neoadjuvant treatment of high-risk early-stage breast cancer: use as an endpoint to support accelerated approval [PDF on the Internet]
.
Silver Spring (MD)
:
U.S. Food and Drug Administration
; 
2012
[cited 2015 Oct 15]. Available from
: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM305501.pdf.
51.
Gianni
L
,
Pienkowski
T
,
Im
Y-H
,
Roman
L
,
Tseng
L-M
,
Liu
M-C
, et al
Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial
.
Lancet Oncol
2012
;
13
:
25
32
.
52.
Choueiri
TK
,
Jacobus
S
,
Bellmunt
J
,
Qu
A
,
Appleman
LJ
,
Tretter
C
, et al
Neoadjuvant dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin with pegfilgrastim support in muscle-invasive urothelial cancer: pathologic, radiologic, and biomarker correlates
.
J Clin Oncol
2014
;
32
:
1889
94
.
53.
Plimack
ER
,
Hoffman-Censits
JH
,
Viterbo
R
,
Trabulsi
EJ
,
Ross
EA
,
Greenberg
RE
, et al
Accelerated methotrexate, vinblastine, doxorubicin, and cisplatin is safe, effective, and efficient neoadjuvant treatment for muscle-invasive bladder cancer: results of a multicenter phase II study with molecular correlates of response and toxicity
.
J Clin Oncol
2014
;
32
:
1895
901
.
54.
Untch
M
,
Fasching
PA
,
Konecny
GE
,
Hasmüller
S
,
Lebeau
A
,
Kreienberg
R
, et al
Pathologic complete response after neoadjuvant chemotherapy plus trastuzumab predicts favorable survival in human epidermal growth factor receptor 2–overexpressing breast cancer: results from the TECHNO trial of the AGO and GBG Study Groups
.
J Clin Oncol
2011
;
29
:
3351
7
.
55.
Betticher
DC
,
Schmitz
S-FH
,
Tötsch
M
,
Hansen
E
,
Joss
C
,
von Briel
C
, et al
Mediastinal lymph node clearance after docetaxel-cisplatin neoadjuvant chemotherapy is prognostic of survival in patients with stage IIIA pN2 non–small cell lung cancer: a multicenter phase II trial
.
J Clin Oncol
2003
;
21
:
1752
9
.
56.
Loibl
S
,
von Minckwitz
G
,
Schneeweiss
A
,
Paepke
S
,
Lehmann
A
,
Rezai
M
, et al
PIK3CA mutations are associated with lower rates of pathologic complete response to anti–human epidermal growth factor receptor 2 (HER2) therapy in primary HER2-overexpressing breast cancer
.
J Clin Oncol
2014
;
32
:
3212
20
.
57.
Wang
C
,
Zhang
J
,
Wang
Y
,
Ouyang
T
,
Li
J
,
Wang
T
, et al
Prevalence of BRCA1 mutations and responses to neoadjuvant chemotherapy among BRCA1 carriers and non-carriers with triple-negative breast cancer
.
Ann Oncol
2015
;
26
:
523
8
.
58.
Salgado
R
,
Denkert
C
,
Campbell
C
,
Savas
P
,
Nucifero
P
,
Aura
C
, et al
Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO trial
.
JAMA Oncol
2015
;
1
:
448
.
59.
Denkert
C
,
von Minckwitz
G
,
Brase
JC
,
Sinn
BV
,
Gade
S
,
Kronenwett
R
, et al
Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2–positive and triple-negative primary breast cancers
.
J Clin Oncol
2015
;
33
:
983
91
.
60.
Brown
JR
,
Wimberly
H
,
Lannin
DR
,
Nixon
C
,
Rimm
DL
,
Bossuyt
V
. 
Multiplexed quantitative analysis of CD3, CD8, and CD20 predicts response to neoadjuvant chemotherapy in breast cancer
.
Clin Cancer Res
2014
;
20
:
5995
6005
.
61.
Mao
Y
,
Qu
Q
,
Zhang
Y
,
Liu
J
,
Chen
X
,
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
:
e115103
.
62.
Allen
EMV
,
Mouw
KW
,
Kim
P
,
Iyer
G
,
Wagle
N
,
Al-Ahmadie
H
, et al
Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma
.
Cancer Discov
2014
;
4
:
1140
53
.
63.
Plimack
ER
,
Dunbrack
RL
,
Brennan
TA
,
Andrake
MD
,
Zhou
Y
,
Serebriiskii
IG
, et al
Defects in DNA repair genes predict response to neoadjuvant cisplatin-based chemotherapy in muscle-invasive bladder cancer
.
Eur Urol
2015
;
68
:
959
67
.
64.
Mariya
T
,
Hirohashi
Y
,
Torigoe
T
,
Asano
T
,
Kuroda
T
,
Yasuda
K
, et al
Prognostic impact of human leukocyte antigen class I expression and association of platinum resistance with immunologic profiles in epithelial ovarian cancer
.
Cancer Immunol Res
2014
;
2
:
1220
9
.