Neoadjuvant therapy for pancreatic cancer: Limitations and advances of response assessment (Review)

  • Authors:
    • Jianwei Xu
    • Hanxiang Zhan
    • Feng Li
    • Sanyuan Hu
    • Lei Wang
  • View Affiliations

  • Published online on: February 18, 2021     https://doi.org/10.3892/or.2021.7977
  • Article Number: 26
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neoadjuvant therapy (NAT) has been widely recommended for managing patients with borderline resectable pancreatic cancer and resectable tumors with high risk factors. Accurate evaluation of the response after NAT is crucial to decide surgery, which then improves the rate of R0 resection and avoids meaningless surgery. The response to NAT is currently evaluated by conventional radiological examination and changes of serum CA19‑9 levels. However, these assessments cannot accurately reflect the response to NAT. This article describes the limitations and advances of NAT response evaluation in pancreatic cancer. The values of some traditional imaging techniques, including positron emission tomography, endoscopic ultrasound, and diffusion weighted magnetic resonance imaging, are discussed, as well as novel imaging modalities or biomarkers, such as radiomics, dual energy computed tomography and liquid biopsy.

Introduction

Neoadjuvant therapy (NAT) has become common practice in managing patients with localized pancreatic cancer (PC). Multimodal NAT strategies have been developed, such as chemotherapy and/or chemoradiotherapy (CRT). The benefits of NAT include increased feasibility of R0 resection, elimination of micrometastases, and identifying aggressive tumors to avoid futile surgery (1). Accurate evaluation of response after NAT is therefore a prerequisite for patients to achieve these benefits from NAT. However, the evaluation of response to NAT is particularly difficult in PC. Consensus and recommendations are still lacking. Conventional radiological examination is one of the most common assessment methods, but this strategy has been identified as unsuitable to evaluate NAT response in PC (2). Another method is monitoring the levels of serum carbohydrate antigen 19-9 (CA19-9); however, the cut-off value of decreased CA19-9 for diagnosing NAT responders remains controversial (3,4). Therefore, it is crucial to identify novel imaging tools or biomarkers. This review presents the challenges and new developments for NAT response evaluation in PC.

Limitations of restage after NAT by conventional radiological images

The decision for further operation after NAT traditionally depends on radiologic imaging, especially contrast enhanced computed tomography (CT). However, radiologic imaging no longer predicts unresectability after NAT for patients with locally advanced PC (LAPC) and borderline resectable PC (BRPC) (5). Response Evaluation Criteria in Solid Tumors (RECIST) are not effective criteria for patients with BRPC (6). Katz et al (6) evaluated the restage values of RECIST for BRPC. Among the 122 patients with BPRC who completed NAT (gemcitabine-based chemotherapy followed by planned chemoradiation or chemoradiation alone) and were restaged, 69% patients had stable disease, 12% had a partial response, and 19% had progressive disease. Although only one patient downstaged to resectable status after NAT, 85 patients (66%) underwent surgical resection. Of that, 81 patients achieved R0 resection. Significant downstaging of PC after NAT is rare; yet, even a minimal radiological response of imaging resulted in a R0 resection of 76.3% (7).

Histological response cannot be reflected by conventional radiological images. The A021101 trial was a prospective, multicenter, single-arm clinical trial, which was designed as patients received modified FOLFIRINOX treatment for four cycles followed by 5.5 weeks of external-beam radiation with capecitabine (orally twice daily) prior to pancreatectomy. In total, 22 patients initiated modified FOLFIRINOX treatment. Results indicated that only 27% of patients with BRPC had a radiologic partial or complete response after NAT, and yet R0 resection and less than 5% residual cancer cells on histopathology were achieved in 93 and 33% of patients, respectively (8). Similarly, Xia et al (7) showed that one-fourth of patients who underwent NAT with gemcitabine-based systemic chemotherapy for BRPC did not have a radiologic response but had an Even's grade IIB or greater pathologic response.

Furthermore, radiologic response was not prognostic with regard to overall survival. Patients with positive radiological response had no differences in overall survival compared with patients without radiological response (6,7).

Reasons for poor evaluation performance of conventional radiological imaging

NAT, especially CRT, causes tumor necrosis, fibrosis, or inflammatory changes, which results in an increase of fibrosis within the lesion and a decrease of cancer cells (9). However, the currently available radiological imaging technology cannot distinguish tissue desmoplastic reactions after NAT and the viable tumor. The sensitivity and specificity of CT and magnetic resonance imaging (MRI) in predicting the presence of viable cancer cells at vessel interfaces after NAT were 71 and 58%, respectively (10). Therefore, the histological response cannot truly be reflected by RECIST that depends on tumor morphology. Tumor size reductions were replaced by intra-tumoral fibrosis (5,11).

Although downstaging or radiological response is not observed, clinically significant cytotoxic activity may occur at the peripheral tumor-vessel interface (6). In the study by Katz et al (6), CT images revealed a close relationship between the tumor and mesenteric vasculature in all patients after NAT, but only one patient had a positive superior mesenteric artery margin. Dholakia et al (12) reported that both tumor volume and degree of tumor-vessel involvement did not significantly change in cases that underwent resection after NAT (induction chemotherapy followed by chemoradiation or upfront chemoradiation). Successful resection is common for patients with BRPC who had no radiographic downstaging or even improvement of tumor-vessel involvement (12).

Artery invasion is a major factor in the staging of PC, which means an aggressive disease with adverse prognosis. The higher morbidity and mortality of arterial resection and reconstruction make surgeons give up radical resections. Of note, conventional radiological images cannot accurately assess artery invasion or infiltration, which might lead to an overestimation of grade of artery invasion, especially for cases that underwent NAT (13). Some cases that were classified as artery invasion or encasement by conventional radiological images could be resected radically without arterial resection and reconstruction by sharp dissection under an artery sheath or on the adventitial layer of a suspected artery (13,14). Hackert and colleagues (13) reported 15 LAPC cases with arterial-sparing resection after NAT with FOLFIRINOX or gemcitabine plus abraxane. All cases were exploration by an ‘artery first’ maneuver. The arterial level of the suspected attachment or encasement was exposed. The adventitial layer was then opened longitudinally and tissues were obtained for frozen section examination. Only cases without viable tumor underwent further resection. R0 resection was achieved in 6 out of 15 patients and the other 9 cases were R1 resections with positive sites located at the peripancreatic soft tissue margins.

Evaluation of NAT response using CA19-9

CA19-9, also known as sialyl Lewis-A, is an indicator of aberrant glycosylation of PC and is widely used as a treatment biomarker. Theoretically, a greater CA19-9 reduction could reflect at least a partial response (7). However, the precise value and cutoff for this diagnosis remain to be elucidated.

First, the exact cut-off value of decreased CA19-9 for diagnosing NAT responders remains controversial. Tsai et al (3) found that normalization of CA19-9 following NAT rather than the magnitude of change was the strongest prognostic marker for long-term survival. Failure to normalize CA19-9 after NAT was associated with a 2.77-fold increased risk of death. Murakami et al (15) showed that patients with arterial contact who achieved normalization of serum CA19-9 or Dupan-2 after NAT are potentially good candidates for tumor resection. By contrast, some studies showed that a decrease in CA19-9 of >50% after NAT treatment was associated with R0 resection rate, histopathologic response, and survival in patients with BRPC (4,16).

Furthermore, pretreatment levels of CA19-9 could influence its application in patient selection. Combs et al (17) showed that pretreatment levels of CA19-9 significantly influenced OS using different cut-off values. With the increase of CA19-9 after NAT, the possibility of surgical resection after NAT decreased from 46% in patients with CA19-9 levels below 90 U/ml to 31% in the group with CA19-9 levels higher than 269 U/ml.

Finally, the value of CA19-9 in patients with negative expression of Lewis antigen (that is critical for CA19-9 biosynthesis) is unclear. Luo et al (18) indicated that CA19-9 could be used as a biomarker in patients with Lewis (−). Authors of that study showed that 8.4% of patients with PC (n=1482) were Lewis (−), but only 41.9% of those cases had CA19-9 values ≤2 U/ml, and 27.4% of cases had elevated levels (>37 U/ml). The area under the receiver operating characteristic curve for CA19-9 as a diagnostic biomarker was 0.842 in Lewis (−) patients with PC, which was similar to that of CA19-9 applied in all of the patients with PC (0.898). Those results mean investigating the values of CA19-9 in monitoring chemotherapy efficiency further.

Advances in the evaluation of response to NAT

Evaluation of responses to NAT by RECIST criteria based on conventional CT/MRI has become challenging, and therefore novel assessment tools are needed. Some conventional imaging techniques have been explored and show potentially favorable values, such as positron emission tomography (PET), endoscopic ultrasound (EUS), and diffusion weighted MRI. Novel imaging modalities or biomarkers have been developed and have shown encouraging results. Advances in the last five years are presented in Table I (3,1954). In addition, considering the limitations of CT that might miss more than 30% of occult metastases in patients with BRPC or LAPC, which would result in an underestimated pretreatment staging and overestimated restaging after NAT treatment, the value of laparoscopic examination before NAT treatment is also discussed in this section.

Table I.

Advances in the evaluation of response to neoadjuvant therapy in the last five years.

Table I.

Advances in the evaluation of response to neoadjuvant therapy in the last five years.

Author/(Refs.)/YearDiseaseNAT regimensToolsIndexResults
Truty et al (28) 2019BRPCFOLFIRINOXPETSUVComplete metabolic response correlated with major pathologic response.
LAPCGEM plus nab-paclitaxel CRT CA19-9CA19-9 response associated with prolonged survival.
Sherman et al (42) 2018LAPCGEM Docetaxel Capecitabine CRTPETSUV; CA 19-9; Tumor size; Degree of vascular involvementNo presenting parameter could predict the success of NAT.
Sakane et al (45) 2017T1-3CRTPETSUVHigher post-treatment SUVpeak and positive MTV/TLG predicted the unfavorable histopathological effects.
N0-1 MTV
M0 TLG
Akita et al (47) 2017RPC BRPCGEM-based CRTPETSUV; Percentage of SUV decline (regression index)The post SUV-max and regression index were related to pathological response.
The sensitivity and specificity of regression index for the detection of Evans grade III/IV were 92.9 and 62.3%.
Mellon et al (50) 2017BRPC LAPCGEM FOLFIRINOX OtherPETSUV CA19-9Tumor regression grade was correlated with CA19-9 or SUVmax.
Nasief et al (19) 2020BRPC LAPCCRTCTDR CA19-9DRF correlated to CA19-9. DRFs-CA19-9 combination predicts treatment response.
Borhani et al (23) 2020RPC BRPCCTTexture analysisPatients with higher MPP at pretreatment CT have favorable histologic response.
Kim et al (34) 2019CRTCTTexture analysisHigher subtracted entropy and lower subtracted grey level co-occurrence matrices entropy are important parameters for prediction of longer OS.
Amer et al (38) 2018Stage I/II/IVCTTumor/parenchyma interfaceChange at the PDAC/parenchyma interface was an early predictor of response to therapy.
Wagner et al (49) 2017BRPC LAPCFOLFIRINOXCTLargest axis; P3A; Arterial/venous involvementThe largest axis/P3A variations were higher in case of complete pathological response.
Klaassen et al (35) 2018PDACMRISix DW-MRI modelsAll models could identify individual treatment effects.
Dalah et al (37) 2018RPC BRPCMRIADCADC values after NAT were correlated with pathological responses.
Trajkovic-Arsic et al (41) 2017LAPC MetastaticGEM-based FOLFIRINOXMRIADCADC could be an early response monitoring tool.
Okada et al (48) 2017BRPCFOLFIRINOX Nab-paclitaxel plus GEM CRTMRIADCThe sensitivity, specificity, accuracy of ADC for discriminating between nonresponders and responders were 100, 75 and 83%.
Ehrlich et al (22) 2019BRPC LAPCEUSEUS-FNA for periarterial soft tissueSensitivity, specificity and accuracy of EUS-FNA for determining resectability were 80, 100 and 92.9%.
Payen et al (20) 2019Brca2-mutant mouse model of PDACCisplatinHarmonic motion imagingStiffnessTissue stiffness decreases when tumors respond successfully to chemotherapy.
Heger et al (26) 2019 UnresectabilityGEM-based FOLFIRINOXBiomarker (blood)CA19-9CA 19-9 levels below 91.8 U/ml as well as a reduction to <40.7% after NAT predict tumor resectability.
Aoki et al (29) 2019Biomarker (blood)CA19-9Decreased CA19-9 levels (≤ 103 U/ml) after NAT predicts a better prognosis.
Michelakos et al (40) 2019BRPC LAPCFOLFIRINOXBiomarker (blood)CA19-9 Tumor sizePreoperative CA 19-9 >100 U/ml and tumor size (>3.0 cm on CT) predicted decreased OS
Tsai et al (3) 2018Localized PCBiomarker (blood)CA19-9Normalization of CA19-9 after NAT, rather than the magnitude of change, is the strongest prognostic marker for long-term survival
Van Veldhuisen et al (36) 2018LAPCFOLFIRINOXBiomarker (blood)CA19-9A CA19-9 decrease ≥30% after NAT was associated with improved survival
Williams et al (54) 2016BRPC LAPCBiomarker (blood)CA19-9Pre-operative CA19-9 decrease could guide treatment duration
Aldakkak et al (52) 2015Localized PCGEM-based CRTBiomarker (blood)CA19-9Pre-treatment CA19-9 could not predict OS.
Normalization of post-treatment CA19-9 in response to NAT was highly prognostic.
Bernard et al (31) 2019Potentially resectable tumorsBiomarker (blood)Liquid biopsy (plasma exoDNA and ctDNA)An increase in exoDNA level after NAT was associated with disease progression.
MAFs ≥5% in exoDNA were a significant predictor of PFS An MAF peak above 1% in exoDNA was associated with radiologic progression.
Gemenetzis et al (32) 2018Biomarker (blood)Liquid biopsy (CTCs)Patients received NAT had lower total CTCs Preoperative numbers of CTCs were predictors of early recurrence in post-NAT patients.
Liang et al (44) 2017Biomarker (blood)Liquid biopsy (tumor-derived extracellular vesicles EphA2-EV)Levels change of plasma EphA2-EV was associated with treatment response.
Murthy et al (24) 20196RPCGEM-based 5FU-basedBiomarker (blood)Systemic immune inflammatory markersElevated post-NAT SII was an independent, negative predictor of OS.
Both Other An 80% reduction in SII predicted a CA 19-9 response after NAT.
Kawai et al (30) 2019BRPC CRTFOLFIRINOXBiomarker (blood)Systemic immune inflammatory markersA low lymphocyte-to-monocyte ratio is useful prognostic factor
Hasegawa et al (53) 2016PDACGEM-based ChemotherapyBiomarker (blood)Systemic immune inflammatory markersPre-treatment NLR was a predictive indicator of pathological response
Felix et al (43) 2018PDAC (Stage I/II–IV)Biomarker (blood)S-TKThe S-TK activity in the NAT group was higher than that in the group not receiving NAT. S-TK activity may be used for monitoring NAT efficacy.
Kuwabara et al (27) 2019PDACBiomarker (tissue)TLOTLO/tumor ratio was an independent predictive prognostic factor.
Tsai et al (33) 2018RPC BRPCBiomarker (tissue)Molecular profiling (6 biomarkers)Molecular profiling may improve the efficacy of chemotherapy
Kurahara et al (39) 2018PDACCRTBiomarker (tissue)GLUT-1Patients with low GLUT-1 expression displayed a better response to NAT.
GLUT-1 expression was significantly increased after NAT treatment.
Yabushita et al (46) 2017BRPCGEM plus S-1 CRTBiomarker (tissue)hENT1; TS; DPDThe presence of three markers was associated with improved partial response rates to NAT
Capello et al (51) 2015LPC, BRPC LAPCFOLFIRINOXBiomarker (tissue)CES2High CES2 expression was associated with longer OS.
Farren et al (21) 2020PDACFOLFIRINOX RadiotherapyBiomarker (tissue)Immunologic microenvironmentNAT modulate tumor, immune, and stromal components of the tumor microenvironment.
Mota et al (25) 2020BRPC LAPCBiomarker (tissue)Immunologic microenvironmentThe degree of antitumor immune remodeling correlates to the degree of histopathologic response to NAT

[i] ADC, apparent diffusion coefficient; BRPC, borderline resectable pancreatic cancer; CES2, carboxylesterase 2; CRT, chemoradiotherapy; CT, computed tomography; CTCs, circulating tumor cells; DPD, dihydropyrimidine dehydrogenase; DR, delta-radiomics; DRF, delta-radiomics features; DW, diffusion-weighted; EUS, endoscopic ultrasound; FNA, fine needle aspiration; GEM, gemcitabine; GLUT-1, glucose transporter type 1; hENT1, equilibrative nucleoside transporter 1; LAPC, locally advanced pancreatic cancer; MAF, mutant allele fraction; MPP, mean positive pixel; MRI, magnetic resonance imaging; MTV, metabolic tumor volume; NAT, neoadjuvant therapy; NLR, neutrophil to lymphocyte ratio; OS, overall survival; P3A, product of the three axes; PC, pancreatic cancer; PDAC, pancreatic ductal adenocarcinoma; PET, positron emission tomography; PFS, progression-free survival; SII, systemic immune-inflammation index; S-TK, serum thymidine kinase 1; SUV, standard uptake value; TLG, total lesion glycolysis; TLO, tertiary lymphoid organs; TS, thymidylate synthase.

18F-fluorodeoxyglucose (FDG)-PET and post-treatment staging

FDG-PET is widely used to diagnose malignant tumors, detect distant metastases, and monitor tumor progression (55), which has also been used in the field of NAT in the assessment of therapeutic efficacy. FDG uptake is closely associated with tumor burden and viability (47), and changes in standard uptake values (SUVs) can reflect the metabolic response of cancer to chemotherapy. Therefore, FDG-PET/CT is used to assess the efficacy of NAT in various solid tumors, such as esophageal adenocarcinoma (56) and triple-negative breast cancer (57). The predictive values of FDG-PET/CT on NAT in PC have been described by only a few reports. A randomized phase III trial examined the feasibility of PET in evaluating the efficacy of nab-paclitaxel plus gemcitabine for the treatment of patients with metastatic PC and showed that patients with higher metabolic response had a longer survival (58). Akita et al (47) showed that FDG-PET could predict the efficacy of neoadjuvant chemoradiotherapy (NACRT) for resectable PC (RPC) and BRPC. The maximum SUVs and tumor size were significantly decreased compared with pretreatment values. However, both the maximum SUV and the percentage of SUV decline (that is, regression index) were significantly related to Evans grade III/IV (59), which grades the pathological response of PC, while only the regression index predicted NACRT efficacy. The AUC of the regression index for the detection of grade III/IV was 0.822 using 50% as the threshold, with a sensitivity and specificity of 92.9 and 62.3%, respectively.

Although declined SUV after NAT showed a good pathological response and prognostic value, there are some limitations of FDG-PET/CT in evaluating the efficacy of NAT using SUV. For example, SUVs are mainly influenced by tumor burden and aggressiveness. However, the physiologic uptake and inflammation in the surrounding non-cancerous tissue can also influence the SUV (55). Many factors cause peripancreatic inflammation, including blockage of the pancreatic duct by the tumor, endoscopic retrograde biliary drainage, radiotherapy, and EUS biopsy (47,55,60,61). Tissues with inflammation show an overlap uptake such as malignant tumors (55), which can lead to falsely elevated SUVs (47). Additionally, the presence of inflammatory cells including macrophages, neutrophils, and fibroblasts can cause high uptake of FDG (55). As we known, PC tissues contain massive stroma, of that, infiltration of inflammatory cells including macrophages, neutrophils, is one of the pathological features of cancer. NAT can induce the infiltration of inflammatory cells and fibrosis even further, and therefore post-therapy SUV might not reflect the real pathological response.

The right timing of FDG-PET/CT examination after NAT is important for improving its predictive value. Ramanathan et al reported that metabolic response rates at week 8 after NAT with nab-paclitaxel plus gemcitabine or gemcitabine alone were similar to the best metabolic response in a randomized phase III trial (58). Akita et al (47) found that 8 weeks after radiotherapy is a good time for FDG-PET/CT examination. No inflammatory changes in the peripancreatic tissues were observed at that time through pathological examination of the resected specimens. A higher specificity (62.9%) was shown compared with previously reported values (47,56).

Compared with traditional RECIST, the metabolic response detected by PET represents a functional measure of tumor response by evaluating metabolic activity and shows a higher accuracy. More patients experienced a metabolic response than a RECIST-defined response in a randomized phase III trial (58). Although the predictive value of PET has some limitations, the correct timing of examination may improve the accuracy. Taken together, these findings indicate that the application of PET in NAT should be further investigated.

Endoscopic ultrasound (EUS)

EUS is rarely used to evaluate NAT response in PC (62). Tumor stiffness on EUS elastography could reflect the abundance of cancer stroma (63), which is the theoretical basis of EUS for response evaluation. For cases that underwent treatment with agents targeting the stroma, such as nab-paclitaxel and gemcitabine, elastography could monitor stromal changes with treatment and show potential value for response evaluation. Alvarez et al (64) indicated that tumor stiffness on EUS elastography was significantly reduced after NAT with nab-paclitaxel and gemcitabine, and the ratio value was decreased from 36 to 18 after treatment. A reduction of tumor stiffness was positively correlated with a decrease of serum CA19-9 levels and resectability.

However, the value of elastography for conventional chemotherapy and radiation therapy was uncertain due to the weak influence of stroma (64). Bettini et al (65) found that the information provided by EUS was not reliable for patient selection for further surgery among those who underwent NAT with radiotherapy combined with 5-fluorouracil and cisplatin chemotherapy.

Diffusion-weighted magnetic resonance imaging (DWI)

DWI is a functional MRI technique that can recognize tissue diffusivity characteristics and perform quantitative and qualitative evaluation of a tumor (66). The utility of DWI in evaluating the responses of PC to NAT has been previously investigated. A pilot study indicated that pretreatment apparent diffusion coefficient (ADC) values differed significantly between responders and non-responders and could be used in the prediction of gemcitabine-based NARCT response (67). The sensitivity, specificity, and accuracy for predicting NAT responders were 100, 75, and 83%, respectively (48). Although DWI had improved performance compared to conventional MRI or CT to assess NAT response in PC (68), the data are limited and obtained from small samples. Further investigation is needed to evaluate the utility of this technique.

Artificial intelligence and radiomics

Artificial intelligence (AI) can be applied to automatically quantify radiographic patterns in medical imaging data (69,70). Compared with human assessment, AI can perform precise volumetric delineation of tumor size, convert intratumoral phenotypic nuances to genotype implications, and recognize complex patterns in images (69,70), which is effective in cancer detection and characterization (71,72). AI can also dynamically monitor a tumor (69), including evaluation of responses to treatment.

Radiomics has been developed based on AI and is widely used in many different precision medicine applications. Unlike traditional radiological images that assess a tumor depending on largely qualitative features, including tumor density, pattern of enhancement, tumor margins, anatomic relationship to the surrounding tissues, and intratumoral composition (69), radiomics is the conversion of digital radiological images into mineable quantitative data (73), including tumor intensity, shape, size or volume, and texture features images are computed and quantified (74,75). Radiomics can reflect a tumor panorama by a non-invasive examination compared with conventional biopsy-based assays that represent only a sample of the tumor (76). This is particularly useful in PC, in which different lesions can exist in different microenvironments due to the excessive stroma and heterogeneous (7779).

Radiomics is beneficial for patient selection. Trebeschi et al (76) found that radiomics may function as a non-invasive biomarker for response to immunotherapy for patients with metastatic melanoma and non-small cell lung cancer and potentially benefit patient stratification in both neoadjuvant and palliative settings. Radiomics can also predict the responses to treatment. A combined intratumoral and peritumoral radiomic feature could successfully predict the pathological complete response of breast cancer to NAT from pretreatment dynamic contrast-enhanced MRI (80). Texture analysis of T2-weighted MR images could predict the efficacy of NACRT in rectal cancer (81). Radiomics based on FDG-PET scans indicated that convolutional neural networks achieve an average 80.7% sensitivity and 81.6% specificity in predicting non-responders of patients with esophageal cancer who underwent NAT (82).

The predictive roles of radiomics in evaluating NAT responses in PC are unclear. Chakraborty et al (83) quantified the heterogeneity of PC by texture analysis and showed that radiomics analysis based on tumor texture could quantify the heterogeneity of PC and optimize patient selection for therapy. In sum, a combination of AI and radiomics presents better values in evaluating NAT responses than traditional radiological images, which is a promising technique in PC.

Dual energy CT (DECT)

DECT enables the differentiation of tumor compositions by simultaneous scanning with different levels of energy and has been used to assess NAT response in gastric and rectal cancer with potential values (84,85). One dilemma of conventional CT/MRI in evaluating NAT response is the difficulty in identifying fibrosis caused by treatment. DECT showed a good performance in the differential diagnosis of PC and chronic mass-forming chronic pancreatitis (86). Considering the features of pathological changes after treatment in PC, quantitative DECT may be useful for monitoring the treatment effects in patients with PC (87). Iodine concentration during DECT could differentiate responders with PC after first-line chemotherapy from non-responders with sensitivity, specificity, and AUC of 97.7, 70.6, and 0.889, respectively (88). Furthermore, low-energy monoenergetic decompositions from DECT may detect an early radiation therapy response because of the increase in soft tissue contrast and the magnitude of radiation-induced changes (89).

New biomarkers

Although serum CA19-9 is generally used to monitor the response to NAT, only moderate predictive values were observed with this marker, and the significance of decreased CA19-9 levels after NAT has not been well clarified (3,29). Serum CA19-9 is also not applicable for patients with negative expression of Lewis antigen (18,90). Therefore, more sensitive and specific biomarkers are needed to assess the response of NAT.

Liquid biopsy includes detection of circulating free deoxyribonucleic acid (cfDNA), circulating tumor cells (CTCs), and exosomes, which displays encouraging value in monitoring the therapeutic response (91). Gemenetzis et al (32) identified two major CTC subtypes, including epithelial CTCs (eCTCs) and epithelial/mesenchymal CTCs (mCTCs). Patients who received NAT had significantly lower levels of both of these CTC subtypes compared with patients with upfront resection. Furthermore, NAT resulted in a significant decrease of CTCs, indicating that CTCs dynamics reflected the response to treatment. Bernard et al (31) showed that an increase in exosome DNA levels after NAT was significantly associated with disease progression. KRAS mutant allele fraction peak in exosome DNA above 1% was significantly associated with radiologic progression. Liang and colleagues reported that levels changes of ephrin type-A receptor 2 in tumor-derived extracellular vesicles (EphA2-EV) were associated with the treatment response. Plasma EphA2-EV levels were significantly decreased in patients with good or partial therapy responses, but not in those with poor response after NAT (44).

Other biomarkers have also been investigated. For patients with BRPC, the presence of three favorable factors (positive expression of human equilibrative nucleoside transporter 1 with negative expression of thymidylate synthase and dihydropyrimidine dehydrogenase) was strongly associated with improved partial response rates to NAT (gemcitabine and S-1 followed by radiotherapy) (46). Serum thymidine kinase (TK) activities in preoperative samples in PC patients who had received NAT were significantly higher than those in patients who had not received NAT. Patients who underwent complete NAT with preoperative serum TK levels <80 Du/l showed a longer OS compared with patients with serum TK levels >80 Du/l (43). Abnormal preoperative glycated haemoglobin A1c (HbA1c) was associated with a 2.74-fold increased odds of metastatic progression during NAT (93). Increased neutrophil-to-lymphocyte ratio after NAT was associated with poor survival after resection of BRPC (93). Tissue expression levels of miRNA-10b are predictive of response to NAT and outcomes in PC (94). Biomarkers from metabolomics analysis are promising. Although few effective metabolic biomarkers have been reported in PC, metabolomics has been used to predict the responses of NAT in breast and rectal cancer (9597).

The above mentioned biomarkers showed potential predictive values in evaluating the responses to NAT. However, almost all studies were preclinical and thus large prospective studies are required to validate these results.

Laparoscopy staging and whether it should be routinely performed

Laparoscopic examination can identify hepatic metastases, peritoneal and serosal implants that may be missed by CT scan then improves the accuracy of cancer staging and avoids unnecessary laparotomy (98). A systematic review and meta-analysis assessed the role of staging laparoscopy in RPC and BRPC and included 1,756 patients with RPC after standard imaging extracted from 12 studies and 242 patients with locally advanced disease from 3 studies. The results showed that 350 of 1,756 cases (20%) were then staged as non-resectable cancer after laparoscopic examination. In addition, 86 of 242 cases (36%) were restaged as metastatic disease (99).

However, the value of laparoscopic staging prior to NAT is unclear. Peng et al (100) evaluated 116 patients with BRPC, including 75 patients who underwent staging diagnostic laparoscopy prior to NAT. There was no difference in overall treatment cost, oncologic treatment, and remaining surgical treatment for patients who underwent laparoscopic examination compared with those who did not. A total of 19 (25%) patients were found with occult abdominal metastases and had a lower overall cost compared with those with negative laparoscopic examination due to avoiding further surgery, radiation, and aggressive chemotherapy (100). Ta et al showed that staging laparoscopy could more accurately select patients for neoadjuvant protocols (99).

The question is which subgroup of patients with PC would benefit from a laparoscopic examination. Slaar et al (101) showed that laparoscopic examination could be performed on selected patients who had a higher chance of metastasis at exploration, such as patients with a tumor ≥3 cm and severe weight loss (≥10 kg) or in patients with a tumor ≥4 cm and moderate weight loss (≥5 kg). National Comprehensive Cancer Network (NCCN) panels recommend laparoscopic examination for resectable PC with high-risk factors (102). Laparoscopic staging should be considered for patients with BRPC prior to NAT. Intraoperative ultrasound is useful during laparoscopic examination to further evaluate the liver, tumor, and vascular involvement.

As an invasive examination, the risks of laparoscopy should be considered, as they may promote trocar-site implantation and peritoneal implant progression. Additionally, there is no clear exploring pathway during laparoscopic staging. Some surgeons merely examine the visible liver and peritoneal surfaces without mobilization and with or without washing for cytology (100). Other surgeons perform extended exploration, including the posterior liver surface, mobilization of the duodenum, evaluation of the proximal jejunal mesentery, and visualization of the lesser sac (103). However, whether extended exploration increases the incidence of tumor implantation remains unknown.

Conclusions and future perspectives

Currently, restaging after NAT in PC mainly depends on conventional radiological examination and changes of serum CA19-9 levels. However, the responses to NAT cannot be accurately reflected by these assessment tools. Several novel imaging modalities or biomarkers have been developed and show promise, but further validation in large samples is needed. In consideration of the histopathological characteristics of PC, novel evaluation techniques that reflect the metabolic response or recognize components of the tumor stroma should be developed in future studies.

Acknowledgements

We would like to thank Liwen Bianji, Edanz Editing China (www.liwenbianji.cn/ac), for editing the English text of a draft of this manuscript.

Funding

This work was supported by the China Postdoctoral Science Foundation (2018M632681), the Natural Science Foundation of China (81502051), and the Shandong Provincial Natural Science Foundation, China (ZR2015HQ010, ZR2017MH032).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

JX, SH, and LW proposed and designed the study. LW and JX wrote the draft. JX, HZ, and FL collected and analyzed the data. All authors contributed to the design and interpretation of the study and to further drafts. HZ, SH, and LW revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Heinrich S and Lang H: Neoadjuvant therapy of pancreatic cancer: Definitions and benefits. Int J Mol Sci. 18:16222017. View Article : Google Scholar

2 

Cassinotto C, Sa-Cunha A and Trillaud H: Radiological evaluation of response to neoadjuvant treatment in pancreatic cancer. Diagn Interv Imaging. 97:1225–1232. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Tsai S, George B, Wittmann D, Ritch PS, Krepline AN, Aldakkak M, Barnes CA, Christians KK, Dua K, Griffin M, et al: Importance of Normalization of CA19-9 levels following neoadjuvant therapy in patients with localized pancreatic cancer. Ann surg. 27:740–747. 2020. View Article : Google Scholar

4 

Boone BA, Steve J, Zenati MS, Hogg ME, Singhi AD, Bartlett DL, Zureikat AH, Bahary N and Zeh HJ III: Serum CA 19-9 response to neoadjuvant therapy is associated with outcome in pancreatic adenocarcinoma. Ann Surg Oncol. 21:4351–4358. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Ferrone CR, Marchegiani G, Hong TS, Ryan DP, Deshpande V, McDonnell EI, Sabbatino F, Santos DD, Allen JN, Blaszkowsky LS, et al: Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann Surg. 261:12–17. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Katz MH, Fleming JB, Bhosale P, Varadhachary G, Lee JE, Wolff R, Wang H, Abbruzzese J, Pisters PW, Vauthey JN, et al: Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators. Cancer. 118:5749–5756. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Xia BT, Fu B, Wang J, Kim Y, Ahmad SA, Dhar VK, Levinsky NC, Hanseman DJ, Habib DA, Wilson GC, et al: Does radiologic response correlate to pathologic response in patients undergoing neoadjuvant therapy for borderline resectable pancreatic malignancy? J Surg Oncol. 115:376–383. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Katz MH, Shi Q, Ahmad SA, Herman JM, Marsh Rde W, Collisson E, Schwartz L, Frankel W, Martin R, Conway W, et al: Preoperative modified FOLFIRINOX treatment followed by capecitabine-based chemoradiation for borderline resectable pancreatic cancer: Alliance for clinical trials in oncology trial A021101. JAMA Surg. 151:e1611372016. View Article : Google Scholar : PubMed/NCBI

9 

Raman SP, Horton KM and Fishman EK: Multimodality imaging of pancreatic cancer-computed tomography, magnetic resonance imaging, and positron emission tomography. Cancer J. 18:511–522. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Donahue TR, Isacoff WH, Hines OJ, Tomlinson JS, Farrell JJ, Bhat YM, Garon E, Clerkin B and Reber HA: Downstaging chemotherapy and alteration in the classic computed tomography/magnetic resonance imaging signs of vascular involvement in patients with pancreaticobiliary malignant tumors: Influence on patient selection for surgery. Arch Surg. 146:836–843. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Hartman DJ and Krasinskas AM: Assessing treatment effect in pancreatic cancer. Arch Pathol Lab Med. 136:100–109. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Dholakia AS, Hacker-Prietz A, Wild AT, Raman SP, Wood LD, Huang P, Laheru DA, Zheng L, De Jesus-Acosta A, Le DT, et al: Resection of borderline resectable pancreatic cancer after neoadjuvant chemoradiation does not depend on improved radiographic appearance of tumor-vessel relationships. J Radiat Oncol. 2:413–425. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Hackert T, Strobel O, Michalski CW, Mihaljevic AL, Mehrabi A, Müller-Stich B, Berchtold C, Ulrich A and Büchler MW: The TRIANGLE operation-radical surgery after neoadjuvant treatment for advanced pancreatic cancer: A single arm observational study. HPB (Oxford). 19:1001–1007. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Klaiber U, Mihaljevic A and Hackert T: Radical pancreatic cancer surgery-with arterial resection. Transl Gastroenterol Hepatol. 4:82019. View Article : Google Scholar : PubMed/NCBI

15 

Murakami Y, Uemura K, Sudo T, Hashimoto Y, Kondo N, Nakagawa N, Okada K, Takahashi S and Sueda T: Prognostic impact of normalization of serum tumor markers following neoadjuvant chemotherapy in patients with borderline resectable pancreatic carcinoma with arterial contact. Cancer Chemother Pharmacol. 79:801–811. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Reni M, Zanon S, Balzano G, Nobile S, Pircher CC, Chiaravalli M, Passoni P, Arcidiacono PG, Nicoletti R, Crippa S, et al: Selecting patients for resection after primary chemotherapy for non-metastatic pancreatic adenocarcinoma. Ann Oncol. 28:2786–2792. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Combs SE, Habermehl D, Kessel KA, Bergmann F, Werner J, Naumann P, Jäger D, Büchler MW and Debus J: Prognostic impact of CA 19-9 on outcome after neoadjuvant chemoradiation in patients with locally advanced pancreatic cancer. Ann Surg Oncol. 21:2801–2807. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Luo G, Fan Z, Cheng H, Jin K, Guo M, Lu Y, Yang C, Fan K, Huang Q, Long J, et al: New observations on the utility of CA19-9 as a biomarker in Lewis negative patients with pancreatic cancer. Pancreatology. 18:971–976. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Nasief H, Hall W, Zheng C, Tsai S, Wang L, Erickson B and Li XA: Improving treatment response prediction for chemoradiation therapy of pancreatic cancer using a combination of delta-radiomics and the clinical biomarker CA19-9. Front Oncol. 9:14642019. View Article : Google Scholar : PubMed/NCBI

20 

Payen T, Oberstein PE, Saharkhiz N, Palermo CF, Sastra SA, Han Y, Nabavizadeh A, Sagalovskiy IR, Orelli B, Rosario V, et al: Harmonic motion imaging of pancreatic tumor stiffness indicates disease state and treatment response. Clin Cancer Res. 26:1297–1308. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Farren MR, Sayegh L, Ware MB, Chen HR, Gong J, Liang Y, Krasinskas A, Maithel SK, Zaidi M, Sarmiento JM, et al: Immunologic alterations in the pancreatic cancer microenvironment of patients treated with neoadjuvant chemotherapy and radiotherapy. JCI Insight. 5:e1303622020. View Article : Google Scholar

22 

Ehrlich D, Ather N, Rahal H, Donahue TR, Hines OJ, Kim S, Sedarat A, Muthusamy VR and Watson R: The Utility of EUS-FNA to determine surgical candidacy in patients with pancreatic cancer after neoadjuvant therapy. J Gastrointest Surg. 24:2807–2813. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Borhani AA, Dewan R, Furlan A, Seiser N, Zureikat AH, Singhi AD, Boone B, Bahary N, Hogg ME, Lotze M, et al: Assessment of response to neoadjuvant therapy using CT texture analysis in patients with resectable and borderline resectable pancreatic ductal adenocarcinoma. AJR Am J Roentgenol. 214:362–369. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Murthy P, Zenati MS, Al Abbas AI, Rieser CJ, Bahary N, Lotze MT, Zeh HJ III, Zureikat AH and Boone BA: Prognostic value of the systemic immune-inflammation index (SII) after neoadjuvant therapy for patients with resected pancreatic cancer. Ann Surg Oncol. 27:898–906. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Mota Reyes C, Teller S, Muckenhuber A, Konukiewitz B, Safak O, Weichert W, Friess H, Ceyhan GO and Demir IE: Neoadjuvant therapy remodels the pancreatic cancer microenvironment via depletion of protumorigenic immune cells. Clin Cancer Res. 26:220–231. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Heger U, Sun H, Hinz U, Klaiber U, Tanaka M, Liu B, Sachsenmaier M, Springfeld C, Michalski CW, Büchler MW and Hackert T: Induction chemotherapy in pancreatic cancer: CA 19-9 may predict resectability and survival. HPB (Oxford). 22:224–232. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Kuwabara S, Tsuchikawa T, Nakamura T, Hatanaka Y, Hatanaka KC, Sasaki K, Ono M, Umemoto K, Suzuki T, Sato O, et al: Prognostic relevance of tertiary lymphoid organs following neoadjuvant chemoradiotherapy in pancreatic ductal adenocarcinoma. Cancer Sci. 110:1853–1862. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Truty MJ, Kendrick ML, Nagorney DM, Smoot RL, Cleary SP, Graham RP, Goenka AH, Hallemeier CL, Haddock MG, Harmsen WS, et al: Factors predicting response, perioperative outcomes, and survival following total neoadjuvant therapy for borderline/locally advanced pancreatic cancer. Ann Surg. 273:341–349. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Aoki S, Motoi F, Murakami Y, Sho M, Satoi S, Honda G, Uemura K, Okada KI, Matsumoto I, Nagai M, et al: Decreased serum carbohydrate antigen 19-9 levels after neoadjuvant therapy predict a better prognosis for patients with pancreatic adenocarcinoma: A multicenter case-control study of 240 patients. BMC Cancer. 19:2522019. View Article : Google Scholar : PubMed/NCBI

30 

Kawai M, Hirono S, Okada KI, Miyazawa M, Shimizu A, Kitahata Y, Kobayashi R, Ueno M, Hayami S, Tanioka K and Yamaue H: Low lymphocyte monocyte ratio after neoadjuvant therapy predicts poor survival after pancreatectomy in patients with borderline resectable pancreatic cancer. Surgery. 165:1151–1160. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Bernard V, Kim DU, San Lucas FA, Castillo J, Allenson K, Mulu FC, Stephens BM, Huang J, Semaan A, Guerrero PA, et al: Circulating nucleic acids are associated with outcomes of patients with pancreatic cancer. Gastroenterology. 156:108–118.e4. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Gemenetzis G, Groot VP, Yu J, Ding D, Teinor JA, Javed AA, Wood LD, Burkhart RA, Cameron JL, Makary MA, et al: Circulating tumor cells dynamics in pancreatic adenocarcinoma correlate with disease status: Results of the prospective CLUSTER Study. Ann Surg. 268:408–420. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Tsai S, Christians KK, George B, Ritch PS, Dua K, Khan A, Mackinnon AC, Tolat P, Ahmad SA, Hall WA, et al: A phase ii clinical trial of molecular profiled neoadjuvant therapy for localized pancreatic ductal adenocarcinoma. Ann Surg. 268:610–619. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Kim BR, Kim JH, Ahn SJ, Joo I, Choi SY, Park SJ and Han JK: CT prediction of resectability and prognosis in patients with pancreatic ductal adenocarcinoma after neoadjuvant treatment using image findings and texture analysis. Eur Radiol. 29:362–372. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Klaassen R, Gurney-Champion OJ, Engelbrecht MRW, Stoker J, Wilmink JW, Besselink MG, Bel A, van Tienhoven G, van Laarhoven HWM and Nederveen AJ: Evaluation of six diffusion-weighted MRI models for assessing effects of neoadjuvant chemoradiation in pancreatic cancer patients. Int J Radiat Oncol Biol Phys. 102:1052–1062. 2018. View Article : Google Scholar : PubMed/NCBI

36 

van Veldhuisen E, Vogel JA, Klompmaker S, Busch OR, van Laarhoven HWM, van Lienden KP, Wilmink JW, Marsman HA and Besselink MG: Added value of CA19-9 response in predicting resectability of locally advanced pancreatic cancer following induction chemotherapy. HPB (Oxford). 20:605–611. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Dalah E, Erickson B, Oshima K, Schott D, Hall WA, Paulson E, Tai A, Knechtges P and Li XA: Correlation of ADC with pathological treatment response for radiation therapy of pancreatic cancer. Transl Oncol. 11:391–398. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Amer AM, Zaid M, Chaudhury B, Elganainy D, Lee Y, Wilke CT, Cloyd J, Wang H, Maitra A, Wolff RA, et al: Imaging-based biomarkers: Changes in the tumor interface of pancreatic ductal adenocarcinoma on computed tomography scans indicate response to cytotoxic therapy. Cancer. 124:1701–1709. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Kurahara H, Maemura K, Mataki Y, Sakoda M, Iino S, Kawasaki Y, Arigami T, Mori S, Kijima Y, Ueno S, et al: Significance of glucose transporter type 1 (GLUT-1) expression in the therapeutic strategy for pancreatic ductal adenocarcinoma. Ann Surg Oncol. 25:1432–1439. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Michelakos T, Pergolini I, Castillo CF, Honselmann KC, Cai L, Deshpande V, Wo JY, Ryan DP, Allen JN, Blaszkowsky LS, et al: Predictors of resectability and survival in patients with borderline and locally advanced pancreatic cancer who underwent neoadjuvant treatment with FOLFIRINOX. Ann Sur. 269:733–740. 2019. View Article : Google Scholar

41 

Trajkovic-Arsic M, Heid I, Steiger K, Gupta A, Fingerle A, Wörner C, Teichmann N, Sengkwawoh-Lueong S, Wenzel P, Beer AJ, et al: Apparent Diffusion Coefficient (ADC) predicts therapy response in pancreatic ductal adenocarcinoma. Sci Rep. 7:170382017. View Article : Google Scholar : PubMed/NCBI

42 

Sherman WH, Hecht E, Leung D and Chu K: Predictors of response and survival in locally advanced adenocarcinoma of the pancreas following neoadjuvant GTX with or without radiation therapy. Oncologist. 23:4–e10. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Felix K, Hinz U, Dobiasch S, Hackert T, Bergmann F, Neumüller M, Gronowitz S, Bergqvist M and Strobel O: Preoperative serum thymidine kinase activity as novel monitoring, prognostic, and predictive biomarker in pancreatic cancer. Pancreas. 47:72–79. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Liang K, Liu F, Fan J, Sun D, Liu C, Lyon CJ, Bernard DW, Li Y, Yokoi K, Katz MH, et al: Nanoplasmonic quantification of tumor-derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat Biomed Eng. 1:00212017. View Article : Google Scholar : PubMed/NCBI

45 

Sakane M, Tatsumi M, Hori M, Onishi H, Tsuboyama T, Nakamoto A, Ota T, Eguchi H, Wakasa K, Hatazawa J and Tomiyama N: Volumetric parameters of 2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography/computed tomography can predict histopathologic treatment response after neoadjuvant chemoradiotherapy in pancreatic adenocarcinoma. Eur J Radiol. 94:64–69. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Yabushita Y, Mori R, Taniguchi K, Matsuyama R, Kumamoto T, Sakamaki K, Kubota K and Endo I: Combined analyses of hENT1, TS, and DPD predict outcomes of borderline-resectable pancreatic cancer. Anticancer Res. 37:2465–2476. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Akita H, Takahashi H, Ohigashi H, Tomokuni A, Kobayashi S, Sugimura K, Miyoshi N, Moon JH, Yasui M, Omori T, et al: FDG-PET predicts treatment efficacy and surgical outcome of pre-operative chemoradiation therapy for resectable and borderline resectable pancreatic cancer. Eur J Surg Oncol. 43:1061–1067. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Okada KI, Hirono S, Kawai M, Miyazawa M, Shimizu A, Kitahata Y, Ueno M, Hayami S, Kojima F and Yamaue H: Value of apparent diffusion coefficient prior to neoadjuvant therapy is a predictor of histologic response in patients with borderline resectable pancreatic carcinoma. J Hepatobiliary Pancreat Sci. 24:161–168. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Wagner M, Antunes C, Pietrasz D, Cassinotto C, Zappa M, Sa Cunha A, Lucidarme O and Bachet JB: CT evaluation after neoadjuvant FOLFIRINOX chemotherapy for borderline and locally advanced pancreatic adenocarcinoma. Eur Radiol. 27:3104–3116. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Mellon EA, Jin WH, Frakes JM, Centeno BA, Strom TJ, Springett GM, Malafa MP, Shridhar R, Hodul PJ and Hoffe SE: Predictors and survival for pathologic tumor response grade in borderline resectable and locally advanced pancreatic cancer treated with induction chemotherapy and neoadjuvant stereotactic body radiotherapy. Acta Oncol. 56:391–397. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Capello M, Lee M, Wang H, Babel I, Katz MH, Fleming JB, Maitra A, Wang H, Tian W, Taguchi A and Hanash SM: Carboxylesterase 2 as a determinant of response to irinotecan and neoadjuvant FOLFIRINOX therapy in pancreatic ductal adenocarcinoma. J Natl Cancer Inst. 107:djv1322015. View Article : Google Scholar : PubMed/NCBI

52 

Aldakkak M, Christians KK, Krepline AN, George B, Ritch PS, Erickson BA, Johnston FM, Evans DB and Tsai S: Pre-treatment carbohydrate antigen 19-9 does not predict the response to neoadjuvant therapy in patients with localized pancreatic cancer. HPB (Oxford). 17:942–952. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Hasegawa S, Eguchi H, Tomokuni A, Tomimaru Y, Asaoka T, Wada H, Hama N, Kawamoto K, Kobayashi S, Marubashi S, et al: Pre-treatment neutrophil to lymphocyte ratio as a predictive marker for pathological response to preoperative chemoradiotherapy in pancreatic cancer. Oncol Lett. 11:1560–1566. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Williams JL, Kadera BE, Nguyen AH, Muthusamy VR, Wainberg ZA, Hines OJ, Reber HA and Donahue TR: CA19-9 normalization during pre-operative treatment predicts longer survival for patients with locally progressed pancreatic cancer. J Gastrointest Surg. 20:1331–1342. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Delbeke D and Martin WH: PET and PET/CT for pancreatic malignancies. Surg Oncol Clin N Am. 19:235–254. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Kukar M, Alnaji RM, Jabi F, Platz TA, Attwood K, Nava H, Ben-David K, Mattson D, Salerno K, Malhotra U, et al: Role of repeat 18F-fluorodeoxyglucose positron emission tomography examination in predicting pathologic response following neoadjuvant chemoradiotherapy for esophageal adenocarcinoma. JAMA Surg. 150:555–562. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Humbert O, Riedinger JM, Charon-Barra C, Berriolo-Riedinger A, Desmoulins I, Lorgis V, Kanoun S, Coutant C, Fumoleau P, Cochet A and Brunotte F: Identification of biomarkers including 18FDG-PET/CT for early prediction of response to neoadjuvant chemotherapy in triple-negative breast cancer. Clin Cancer Res. 21:5460–5468. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Ramanathan RK, Goldstein D, Korn RL, Arena F, Moore M, Siena S, Teixeira L, Tabernero J, Van Laethem JL, Liu H, et al: Positron emission tomography response evaluation from a randomized phase III trial of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone for patients with metastatic adenocarcinoma of the pancreas. Ann Oncol. 27:648–653. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Evans DB, Rich TA, Byrd DR, Cleary KR, Connelly JH, Levin B, Charnsangavej C, Fenoglio CJ and Ames FC: Preoperative chemoradiation and pancreaticoduodenectomy for adenocarcinoma of the pancreas. Arch Surg. 127:1335–1339. 1992. View Article : Google Scholar : PubMed/NCBI

60 

Miyata T, Kamata K and Takenaka M: Endoscopic ultrasonography-guided transenteric pancreatic duct drainage without cautery for obstructive pancreatitis as a result of ampullary carcinoma. Dig Endosc. 30:403–404. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Rosenthal MH, Lee A and Jajoo K: Imaging and endoscopic approaches to pancreatic cancer. Hematol Oncol Clin North Am. 29:675–699. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Barreto SG, Loveday B, Windsor JA and Pandanaboyana S: Detecting tumour response and predicting resectability after neoadjuvant therapy for borderline resectable and locally advanced pancreatic cancer. ANZ J Surg. 89:481–487. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Shi S, Liang C, Xu J, Meng Q, Hua J, Yang X, Ni Q and Yu X: The strain ratio as obtained by endoscopic ultrasonography elastography correlates with the stroma proportion and the prognosis of local pancreatic cancer. Ann Surg. 271:559–565. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Alvarez R, Musteanu M, Garcia-Garcia E, Lopez-Casas PP, Megias D, Guerra C, Muñoz M, Quijano Y, Cubillo A, Rodriguez-Pascual J, et al: Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br J Cancer. 109:926–933. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Bettini N, Moutardier V, Turrini O, Bories E, Monges G, Giovannini M and Delpero JR: Preoperative locoregional re-evaluation by endoscopic ultrasound in pancreatic ductal adenocarcinoma after neoadjuvant chemoradiation. Gastroenterol Clin Biol. 29:659–663. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Baliyan V, Kordbacheh H, Parakh A and Kambadakone A: Response assessment in pancreatic ductal adenocarcinoma: Role of imaging. Abdom Radiol (NY). 43:435–444. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Cuneo KC, Chenevert TL, Ben-Josef E, Feng MU, Greenson JK, Hussain HK, Simeone DM, Schipper MJ, Anderson MA, Zalupski MM, et al: A pilot study of diffusion-weighted MRI in patients undergoing neoadjuvant chemoradiation for pancreatic cancer. Transl Oncol. 7:644–649. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Granata V, Fusco R, Setola SV, Piccirillo M, Leongito M, Palaia R, Granata F, Lastoria S, Izzo F and Petrillo A: Early radiological assessment of locally advanced pancreatic cancer treated with electrochemotherapy. World J Gastroenterol. 23:4767–4778. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, et al: Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin. 69:127–157. 2019.PubMed/NCBI

70 

Fazal MI, Patel ME, Tye J and Gupta Y: The past, present and future role of artificial intelligence in imaging. Eur J Radiol. 105:246–250. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Rajkomar A, Dean J and Kohane I: Machine learning in medicine. N Engl J Med. 380:1347–1358. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Kantarjian H and Yu PP: Artificial intelligence, big data, and cancer. JAMA Oncology. 1:573–574. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Chu LC, Goggins MG and Fishman EK: Diagnosis and detection of pancreatic cancer. Cancer J. 23:333–342. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Liu Z, Wang S, Dong D, Wei J, Fang C, Zhou X, Sun K, Li L, Li B, Wang M and Tian J: The applications of radiomics in precision diagnosis and treatment of oncology: Opportunities and challenges. Theranostics. 9:1303–1322. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Parekh VS and Jacobs MA: Deep learning and radiomics in precision medicine. Expert Rev Precis Med Drug Dev. 4:59–72. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Trebeschi S, Drago SG, Birkbak NJ, Kurilova I, Cǎlin AM, Delli Pizzi A, Lalezari F, Lambregts DMJ, Rohaan MW, Parmar C, et al: Predicting response to cancer immunotherapy using non-invasive radiomic biomarkers. Ann Oncol. 30:998–1004. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Juiz NA, Iovanna J and Dusetti N: Pancreatic cancer heterogeneity can be explained beyond the genome. Front Oncol. 9:2462019. View Article : Google Scholar : PubMed/NCBI

78 

Neesse A, Algul H, Tuveson DA and Gress TM: Stromal biology and therapy in pancreatic cancer: A changing paradigm. Gut. 64:1476–1484. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Dougan SK: The pancreatic cancer microenvironment. Cancer J. 23:321–325. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Braman NM, Etesami M, Prasanna P, Dubchuk C, Gilmore H, Tiwari P, Plecha D and Madabhushi A: Intratumoral and peritumoral radiomics for the pretreatment prediction of pathological complete response to neoadjuvant chemotherapy based on breast DCE-MRI. Breast Cancer Res. 19:572017. View Article : Google Scholar : PubMed/NCBI

81 

Shu Z, Fang S, Ye Q, Mao D, Cao H, Pang P and Gong X: Prediction of efficacy of neoadjuvant chemoradiotherapy for rectal cancer: The value of texture analysis of magnetic resonance images. Abdom Radiol (NY). 44:3755–3784. 2019.PubMed/NCBI

82 

Ypsilantis PP, Siddique M, Sohn HM, Davies A, Cook G, Goh V and Montana G: Predicting response to neoadjuvant chemotherapy with PET imaging using convolutional neural networks. PLoS One. 10:e01370362015. View Article : Google Scholar : PubMed/NCBI

83 

Chakraborty J, Langdon-Embry L, Cunanan KM, Escalon JG, Allen PJ, Lowery MA, O'Reilly EM, Gönen M, Do RG and Simpson AL: Preliminary study of tumor heterogeneity in imaging predicts two year survival in pancreatic cancer patients. PLoS One. 12:e01880222017. View Article : Google Scholar : PubMed/NCBI

84 

Al-Najami I, Drue HC, Steele R and Baatrup G: Dual energy CT-a possible new method to assess regression of rectal cancers after neoadjuvant treatment. J Surg Oncol. 116:984–988. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Yang L, Li Y, Shi GF, Zhou T and Tan BB: The concentration of iodine in perigastric adipose tissue: A novel index for the assessment of serosal invasion in patients with gastric cancer after neoadjuvant chemotherapy. Digestion. 98:87–94. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Yin Q, Zou X, Zai X, Wu Z, Wu Q, Jiang X, Chen H and Miao F: Pancreatic ductal adenocarcinoma and chronic mass-forming pancreatitis: Differentiation with dual-energy MDCT in spectral imaging mode. Eur J Radiol. 84:2470–2476. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Kawamoto S, Fuld MK, Laheru D, Huang P and Fishman EK: Assessment of iodine uptake by pancreatic cancer following chemotherapy using dual-energy CT. Abdom Radiol (NY). 43:445–456. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Noda Y, Goshima S, Miyoshi T, Kawada H, Kawai N, Tanahashi Y and Matsuo M: Assessing chemotherapeutic response in pancreatic ductal adenocarcinoma: Histogram analysis of iodine concentration and CT number in single-source dual-energy CT. AJR Am J Roentgenol. 211:1221–1226. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Noid G, Tai A, Schott D, Mistry N, Liu Y, Gilat-Schmidt T, Robbins JR and Li XA: Technical Note: Enhancing soft tissue contrast and radiation-induced image changes with dual-energy CT for radiation therapy. Med Phys. Jul 4–2018.(Epub ahead of print). doi: 10.1002/mp.13083. View Article : Google Scholar : PubMed/NCBI

90 

Luo G, Liu C, Guo M, Cheng H, Lu Y, Jin K, Liu L, Long J, Xu J, Lu R, et al: Potential biomarkers in lewis negative patients with pancreatic cancer. Ann Surg. 265:800–805. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Zhou B, Xu JW, Cheng YG, Gao JY, Hu SY, Wang L and Zhan HX: Early detection of pancreatic cancer: Where are we now and where are we going? Int J Cancer. 141:231–241. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Rajamanickam ES, Christians KK, Aldakkak M, Krepline AN, Ritch PS, George B, Erickson BA, Foley WD, Aburajab M, Evans DB and Tsai S: Poor Glycemic control is associated with failure to complete neoadjuvant therapy and surgery in patients with localized pancreatic cancer. J Gastrointest Surg. 21:496–505. 2017. View Article : Google Scholar : PubMed/NCBI

93 

Glazer ES, Rashid OM, Pimiento JM, Hodul PJ and Malafa MP: Increased neutrophil-to-lymphocyte ratio after neoadjuvant therapy is associated with worse survival after resection of borderline resectable pancreatic ductal adenocarcinoma. Surgery. 160:1288–1293. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Preis M, Gardner TB, Gordon SR, Pipas JM, Mackenzie TA, Klein EE, Longnecker DS, Gutmann EJ, Sempere LF and Korc M: MicroRNA-10b expression correlates with response to neoadjuvant therapy and survival in pancreatic ductal adenocarcinoma. Clin Cancer Res. 17:5812–5821. 2011. View Article : Google Scholar : PubMed/NCBI

95 

Battini S, Faitot F, Imperiale A, Cicek AE, Heimburger C, Averous G, Bachellier P and Namer IJ: Metabolomics approaches in pancreatic adenocarcinoma: Tumor metabolism profiling predicts clinical outcome of patients. BMC Med. 15:562017. View Article : Google Scholar : PubMed/NCBI

96 

Jia H, Shen X, Guan Y, Xu M, Tu J, Mo M, Xie L, Yuan J, Zhang Z, Cai S, et al: Predicting the pathological response to neoadjuvant chemoradiation using untargeted metabolomics in locally advanced rectal cancer. Radiother Oncol. 128:548–556. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Wei S, Liu L, Zhang J, Bowers J, Gowda GA, Seeger H, Fehm T, Neubauer HJ, Vogel U, Clare SE and Raftery D: Metabolomics approach for predicting response to neoadjuvant chemotherapy for breast cancer. Mol Oncol. 7:297–307. 2013. View Article : Google Scholar : PubMed/NCBI

98 

Allen VB, Gurusamy KS, Takwoingi Y, Kalia A and Davidson BR: Diagnostic accuracy of laparoscopy following computed tomography (CT) scanning for assessing the resectability with curative intent in pancreatic and periampullary cancer. Cochrane Database Syst Rev. 7:Cd0093232016.PubMed/NCBI

99 

Ta R, O'Connor DB, Sulistijo A, Chung B and Conlon KC: The role of staging laparoscopy in resectable and borderline resectable pancreatic cancer: A systematic review and meta-analysis. Dig Surg. 36:251–260. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Peng JS, Mino J, Monteiro R, Morris-Stiff G, Ali NS, Wey J, El-Hayek KM, Walsh RM and Chalikonda S: Diagnostic laparoscopy prior to neoadjuvant therapy in pancreatic cancer is high yield: An analysis of outcomes and costs. J Gastrointest Surg. 21:1420–1427. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Slaar A, Eshuis WJ, van der Gaag NA, Nio CY, Busch OR, van Gulik TM, Reitsma JB and Gouma DJ: Predicting distant metastasis in patients with suspected pancreatic and periampullary tumors for selective use of staging laparoscopy. World J Surg. 35:2528–2534. 2011. View Article : Google Scholar : PubMed/NCBI

102 

NCCN, . NCCN Clinical Practice Guidelines in Oncology: Pancreatic Adenocarcinoma (version 1.2019). http://www.nccn.org

103 

Schnelldorfer T, Gagnon AI, Birkett RT, Reynolds G, Murphy KM and Jenkins RL: Staging laparoscopy in pancreatic cancer: A potential role for advanced laparoscopic techniques. J Am Coll Surg. 218:1201–1206. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2021
Volume 45 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu J, Zhan H, Li F, Hu S and Wang L: Neoadjuvant therapy for pancreatic cancer: Limitations and advances of response assessment (Review). Oncol Rep 45: 26, 2021
APA
Xu, J., Zhan, H., Li, F., Hu, S., & Wang, L. (2021). Neoadjuvant therapy for pancreatic cancer: Limitations and advances of response assessment (Review). Oncology Reports, 45, 26. https://doi.org/10.3892/or.2021.7977
MLA
Xu, J., Zhan, H., Li, F., Hu, S., Wang, L."Neoadjuvant therapy for pancreatic cancer: Limitations and advances of response assessment (Review)". Oncology Reports 45.4 (2021): 26.
Chicago
Xu, J., Zhan, H., Li, F., Hu, S., Wang, L."Neoadjuvant therapy for pancreatic cancer: Limitations and advances of response assessment (Review)". Oncology Reports 45, no. 4 (2021): 26. https://doi.org/10.3892/or.2021.7977