Next Article in Journal
Targeted-Lymphoma Drug Delivery System Based on the Sgc8-c Aptamer
Next Article in Special Issue
Predictive and Prognostic Value of Oncogene Mutations and Microsatellite Instability in Locally-Advanced Rectal Cancer Treated with Neoadjuvant Radiation-Based Therapy: A Systematic Review and Meta-Analysis
Previous Article in Journal
Immune-Activated B Cells Are Dominant in Prostate Cancer
Previous Article in Special Issue
Development of a Patient Decision Aid for Rectal Cancer Patients with Clinical Complete Response after Neo-Adjuvant Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Integrated Intensified Chemoradiation in the Setting of Total Neoadjuvant Therapy (TNT) in Patients with Locally Advanced Rectal Cancer: A Retrospective Single-Arm Study on Feasibility and Efficacy

by
Maria Chiara Lo Greco
1,*,
Madalina La Rocca
1,
Giorgia Marano
1,
Irene Finocchiaro
1,
Rocco Luca Emanuele Liardo
2,
Roberto Milazzotto
2,
Grazia Acquaviva
3,
Antonello Basile
4,5,
Stefano Palmucci
4,5,
Pietro Valerio Foti
4,5,
Stefano Pergolizzi
1,6,
Antonio Pontoriero
1,6,
Silvana Parisi
6 and
Corrado Spatola
2,5,*
1
Department Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali, Università di Messina, 98122 Messina, Italy
2
U.O.S.D. Radioterapia Oncologica, A.O.U. Policlinico “G. Rodolico-San Marco” Catania, Via Santa Sofia 78, 95123 Catania, Italy
3
U.O.C. Radioterapia, A.O.E. Cannizzaro, 95126 Catania, Italy
4
U.O. Radiologia I, A.O.U. Policlinico “G. Rodolico-San Marco”, Via Santa Sofia 78, 95123 Catania, Italy
5
Department Scienze Mediche, Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, Università di Catania, 95123 Catania, Italy
6
U.O.C. Radioterapia Oncologica, A.O.U. Policlinico “G. Martino” Messina, Via Consolare Valeria 1, 98125 Messina, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(3), 921; https://doi.org/10.3390/cancers15030921
Submission received: 28 November 2022 / Revised: 16 January 2023 / Accepted: 30 January 2023 / Published: 1 February 2023
(This article belongs to the Special Issue Advances in Radiotherapy and Prognosis of Rectal Cancer)

Abstract

:

Simple Summary

Based on literature data suggesting promising advantages of total neoadjuvant therapy (TNT) in patients with locally advanced rectal cancer, we performed a retrospective single-arm, single-center study on 45 patients affected by histologically and radiologically proven locally advanced rectal cancer, with the aim of analyzing the feasibility and short-term efficacy of an integrated intensified treatment, including induction chemotherapy, concurrent chemoradiation with long-course radiotherapy, and concomitant boost and consolidation chemotherapy. At a median follow-up of 30 months, this strategy has shown to be feasible and effective in terms of pathological complete response (pCR) and short-term disease-free survival (DFS).

Abstract

While surgery is considered the main treatment for early-stage rectal cancer, locally advanced rectal cancer needs to be handled with a multidisciplinary approach. Based on literature data suggesting promising advantages of total neoadjuvant therapy (TNT), we performed a retrospective, single-arm, single-center study on 45 patients affected by histologically and radiologically proven locally advanced rectal cancer, with the aim of analyzing the feasibility and short-term efficacy of an integrated intensified treatment in the setting of TNT. Each analyzed patient performed three cycles of FOLFOX4 or De Gramont induction chemotherapy (iCT), followed by concurrent chemoradiotherapy (CRT) with long course radiotherapy (LCRT) plus concomitant boost and continuous 5-FU infusion, followed by three cycles of FOLFOX4 or De Gramont consolidation chemotherapy (conCT) and then surgery with total mesorectal excision. At a median follow-up of 30 months, this strategy has shown to be feasible and effective in terms of pathological complete response (pCR) and short-term disease-free survival (DFS).

1. Introduction

Colorectal cancer is the third most common cancer in western countries and the second leading cause of cancer death in the world [1].
During the last few decades, the outcome for patients with rectal cancer has significantly improved in many high-income countries, due to the development of various treatment strategies.
In the early 1980s, a better understanding of rectal surgical oncology led to the introduction of a new operative technique, called total mesorectal excision (TME), a specific surgical approach consisting in the complete removal of the entire circumferential perirectal tissue envelope. Due to the clinical practice of TME, the 5-year survival improved from 45–50% to 75%, local recurrence rates decreased from 30% to 5–8%, the sphincter preservation for mid- and lower rectal cancer increased by 20%, and impotence and bladder dysfunction rates declined from 50–85% to 15% [2,3].
In the following years, multiple trials investigating the role of neoadjuvant therapies in the management of locally advanced rectal cancer (LARC) assessed the advantages of delivering preoperative chemoradiotherapy (CRT) to improve local control rates and increase sphincter preservation rates in patients with low-lying tumors [4,5,6].
Despite the current standard of care is combined-modality therapy withneoadjuvant CRT, surgery with TME and adjuvant chemotherapy, distant metastasis remains the major reason for treatment failure. In the aim to reduce the risk of distant recurrence, several studies investigated the potential advantage of a novel approach, termed total neoadjuvant therapy (TNT), which consists of delivering systemic chemotherapy in the neoadjuvant setting, either before or after CRT. Early results have demonstrated high rates of completion and tolerability, pathological complete response (pCR), sphincter-saving surgery, and R0 resection. Moreover, the current pooled data has shown TNT benefit in the rate of disease-free survival (DFS) and 3-year overall survival (OS) [7].
Based on literature data suggesting promising advantages of TNT strategy, we performed a retrospective, single-arm, single-center study with the aim of analyzing the feasibility and toxicity rates of an integrated intensified treatment in the setting of TNT. Moreover, we investigated short-term TNT efficacy, in terms of pCR rates and DFS.

2. Materials and Methods

At the University of Catania, we retrospectively analyzed 45 patients affected by histologically and radiologically proven locally advanced rectal cancer, from 2017 to 2021.
After a careful history and physical examination, including digital rectal exam, all patients underwent an endoscopic examination with rigid sigmoidoscopy both to measure the distance from the lesion to the anal verge and to pathologically confirm rectal cancer diagnosis.
All patients underwent pelvic magnetic resonance imaging (MRI) to determine local tumor extension and nodal status and computed tomography (CT) of the brain, chest, abdomen, and pelvis to identify metastatic lesions. Then, they were all staged according to UICC TNM staging (8th edition) classification [8].
We selected 45 patients according to the following inclusion criteria: tumor location within 15 cm of the anal verge, histology of adenocarcinoma, locally advanced clinical stage with absence of metastasis, written informed consent, and performance status > 80% (Karnofsky scale).
All patients were candidates for an integrated intensified treatment in the setting of TNT, which consisted of three cycles of FOLFOX4 or De Gramont induction chemotherapy (iCT), followed by concurrent CRT with long course radiotherapy (LCRT) plus concomitant boost and continuous 5-fluorouracil (5-FU) infusion, followed by three cycles of FOLFOX4 or De Gramont consolidation chemotherapy (conCT) and then surgery with TME.
After surgery, all patients underwent trimestral follow-up for the first year, and then semestral follow-up from the second year onward. At each visit, all patients were clinical and hematological evaluated. Moreover, they underwent semestral instrumental evaluation with CT scan (brain, chest, abdomen, and pelvis) or colonscopy.
To determinate the feasibility and toxicity rates, all patients were clinical and hematological evaluated both during iCT, CRT and conCT. Toxicity data were collected according to Common Terminology Criteria for Adverse Events (CTCAE v4.0) [9].
To determine short-term efficacy, we analyzed pCR rates after surgery, approximately 8 weeks after the end of TNT. Then we assessed DFS, at a median follow-up of 30 months.

2.1. Patients’ Data

From 2017 to 2021, a total of 45 patients were selected to be analyzed in this study; 30 patients were males and 15 were females. The patients’ ages ranged between 43 and 85 years (median = 70 years). Clinical presentation was mostly characterized by different onset symptoms, such as rectal bleeding and abdominal pain (44.4%), change in bowel habits or irregular alvus (40%), tenesmus (37.7%), and sub-obstruction symptoms (26.6%).
After endoscopic examination with rigid sigmoidoscopy, 18 patients were found to be affected by low rectal cancer (<6 cm), 22 patients by middle rectal cancer (6.1–10 cm), and 5 patients by high rectal cancer (>10 cm).
At pretreatment TNM staging (cTNM), 7 patients were cT2 cN+, 8 patients were cT3-T4 cN0, 30 patients were cT3-4 cN+. The characteristics of the patients are listed in Table 1.

2.2. Induction, Concurrent and Consolidation Chemotherapy Schemes

Prior to the start of iCT, all patients received a peripherally inserted central catheter (PICC), and then, to avoid severe adverse events related to fluoropyrimidine treatment, the activity of the DPD enzyme was tested [10]. All patients were found to have wild-type DPYD. Before every chemotherapy infusion, premedication with dexamethasone (8 mg) and ondansetron (8 mg) was administered.
ICT was performed according to the following schemes:
-
FOLFOX4 regimen, based on Oxaliplatin 85 mg/m2 intravenous (IV) infusion (day 1) and leucovorin 200 mg/m2 IV infusion administered concurrently over 120 min, followed by 5-FU 400 mg/m2 IV bolus, followed by 5-FU 600 mg/m2 IV infusion over 22 h (day 1 and 2) every two weeks. The FOLFOX4 induction regimen was administered to 35 patients.
-
De Gramont regimen, based on leucovorin 200 mg/m2 IV infusion over 120 min (day 1 and 2) followed by 5-FU 400 mg/m2 IV bolus (day 1 and 2), followed by 5-FU 600 mg/m2 IV infusion over 22 h (day 1 and 2), leucovorin 400 mg/m2, every two weeks. The De Gramont induction regimen was administered to 10 patients.
The choice of which chemotherapy schedule to perform was tailored based on patients’ characteristics. The De Gramont regimen was preferred in very elderly patients or patients with two or more comorbidities, to avoid severe toxicity related to oxaliplatin.
Afterwards, each one of the selected patients received CRT with LCRT concurrent with fluoropyrimidine based chemotherapy.
Concurrent chemotherapy was performed according to the following schemes:
-
5-FU continuous infusion regimen, based on 5 FU 225 mg/m2 IV infusion over 24 h daily (1–5 or 1–7), every week for five weeks. The 5-FU continuous infusion regimen was administered to all patients.
About two weeks after the last radiotherapy fraction, conCT was performed following the same schemes as iCT.
-
The FOLFOX4 consolidation regimen was administered to 32 patients.
-
The De Gramont consolidation regimen was administered to 13 patients.

2.3. Radiotherapy Treatment Planning and Delivery

To design a treatment plan unique for each patient’s anatomy, after an adequate bowel preparation, every patient underwent a radiotherapy simulation process through an abdomen-pelvis CT scan without contrast enhancement.
Since the patient’s position must be the same for both initial simulation and subsequent treatment, during this phase an optimal set-up was needed: all patients were placed in the prone position using a belly board device, used to displace the small bowels out of the treatment fields.
CT images were acquired with 3–5 mm slice spacing, from L1 to femur half, then data were analyzed using treatment planning software (XIO) for target volume definition and dose solutions, according to our institutional protocol [11].
During the contour delineation phase, a gross tumor volume (GTV) was outlined, including the primary tumor and enlarged regional nodes, and the clinical target volume (CTV) was outlined including the GTV, rectum, mesorectum and posterior pelvic subregions. The mesorectum, presacral, obturator, and internal iliac lymph nodes were included in all patients’ treatment volumes, while the external iliac lymph nodes were included only if clinically positive or in case of T4 tumor.
A planning target volume (PTV) was obtained, giving an expansion of approximately 5 mm (three dimensional) to CTV, to account for daily set-up error and organ motion.
The organ at risk (OARs) were bowel (Dax < 55 Gy), bladder (V50 60%, V60 50%), femoral heads (V50 60%) and anal canal (Dmax < 55 Gy).
Radiation therapy was delivered by means of a Siemens ONCOR linear accelerator using a 3D-conformal technique (3D-CRT) at 6–15 MV or a static step-and-shot intensity-modulated technique (IMRT). IMRT was preferred to avoid intestinal adhesions in patients with previous abdominal surgery and to avoid a high dose to the small bowel with a 3DCRT treatment plan (V15 > 120 cc or V45 > 195 cc of small bowel) [12].
During the entire course of radiotherapy treatment, patients were set up daily by using one sagittal and two lateral tattoos and lasers. A megavoltage cone-beam computer tomography image-guided radiotherapy (MV CBCT IGRT) system was used two times a week to check patients’ setups.
Different fractionation schedules were used, including conventionally fractionated radiation therapy (CFRT) or hypofractionated radiation therapy (HFRT). Total dose and dose per fraction were established considering an α/β ratio of 5.06 Gy, as suggested by literature data [13].
-
CFRT consisted of a total dose of 45–46 Gy in 23–25 fractions (1.8–2 Gy/day) delivered to PTV. During the last week of treatment, a boost of 8–9 Gy in 4–5 fractions (1.8–2 Gy/day) was delivered to GTV, so that this volume received a total dose of 54 Gy.
CRFT was administered to 33 patients.
-
HFRT consisted of a total dose of 40–42.75 Gy in 16–19 fractions (2.25–2.5 Gy/day) delivered to PTV. During the last week of treatment, a boost of 9–10 Gy in 4 fractions (2.25–2.5 Gy/day) was delivered to GTV. So that this volume received a total dose of 50–51.75 Gy.
HFRT was administered to 12 patients.
The characteristics of treatment are listed in Figure 1.

2.4. Patients’ Evaluation

To determine the feasibility and toxicity rates of the TNT approach, before every iCT and conCT cycle and once a week during the entire course of CRT, all patients were clinical and hematological evaluated to identify adverse events. Toxicities were defined following the Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0.
All patients underwent pelvic MRI and CT scans of the brain, chest, abdomen, and pelvis at the time of diagnosis to be staged, and again about 6–7 weeks after the end of TNT, to analyze radiological response. No re-evaluation imaging was planned at the end of iCT phase.
Radiological response was evaluated according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1) [14].
To improve the diagnostic performance of MRI to evaluate the tumor response to TNT, high-resolution T2-weighted and diffusion-weighted imaging (DWI), with qualitative and quantitative evaluation through the measurement of tumors; apparent diffusion coefficient (ADC) were used, according to our institutional protocol [15,16].
After surgery, which was performed approximately 8 weeks after the end of TNT, pathological complete response rates were analyzed.
The imaging and the post-operative staging were then compared to evaluate the correspondence between the clinical and pathological assessments.
To assess the benefits in terms of disease-free survival, after surgery, all patients underwent trimestral follow-up for the first year, and semestral follow-up from the second year after surgery onward. Median follow-up was 30 months.

3. Results

3.1. Feasibility and Toxicity of Integrated Intensified TNT

During iCT and conCT, the majority of patients presented mild adverse events related to oxaliplatin or fluoropyrimidines (Figure 1). Due to poor clinical tolerance, we decided to switch three patients receiving FOLFOX4 iCT to De Gramont conCT.
Among patients performing iCT or conCT with FOLFOX4 regimen, five patients (11.1%) manifested G1 anemia and decreased platelet count; three patients (6.6%) experienced G1 hypertransaminasemia, lymphopenia and neutropenia; four patients (8.8%) experienced constipation and fatigue; and three patients (6.6%) experienced fatigue, nausea, and vomiting. ICT dose modification was performed in four patients (8.8%) due to G2 G.I. toxicity (nausea and vomiting, diarrhoea). ICT interruption (1–2 weeks) was performed in two patients (4.4%) due to G2 anemia and decreased platelet count. Of all patients receiving iCT and conCT with the FOLFOX4 regimen, five patients (11.1%) experienced G1 peripheral neuropathy.
Among patients receiving iCT or conCT with the De Gramont regimen, three patients (6.6%) manifested G1 anemia and decreased platelet count. ICT dose modification was performed in two patients (4.4%) due to G2 decreased platelet count.
All the chemoradiotherapy schemes were well-tolerated in the majority of patients.
During the course of CRT, five patients (11.1%) and three patients (6.6%) manifested G1 and G2 radiation dermatitis, respectively; three patients (6.6%) undergoing CFRT, and 1 patient (2.2%) undergoing HFRT, had their treatment interrupted (from 2 to 7 days) due to G3 diarrhea and proctitis. Moreover, four patients (8.8%) manifested G1 anemia and decreased platelet count.
The majority of patients (66.6%) experienced symptomatic relief during or after TNT. No significant differences between the different chemotherapy regimens or radiotherapy schedules were detected.
Regarding chronic toxicities, after 18 months, only three patients (6.6%) experienced peripheral neuropathy, and four patients (8.8%) experienced G1 proctitis.

3.2. Efficacy of Integrated Intensified TNT

About 6–7 weeks after the end of TNT, all patients underwent pelvic MRI and CT scans of the brain, chest, abdomen, and pelvis. Clinical TNM after neoadjuvant therapies (ycTNM) was evaluated according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1) and then compared to pathological TNM (ypTNM) after surgery (as shown in Figure 2):
-
Partial response (ycPR) was reported in 24 patients (53.3%).
-
Complete response (ycCR)was reported in 8 patients (17.7%).
-
Stable disease (ycSD) was reported in 13 patients (28.8%).
Surgery was performed approximately eight weeks after the end of TNT. A low anterior resection with sphincter preservation was performed in 37 patients, and a temporary diverting loop ileostomy was performed in 25 patients. Miles abdomino-perineal resection was performed in eight patients. The results of all patients (100%) were R0.
The response obtained after TNT allowed surgeons to perform sphincter-saving surgery in 10 out of 18 patients initially selected for abdomino-perineal resection (distal tumour location < 6 cm).
The only post-surgical complications observed were perianastomotic fistula and intestinal obstruction (two patients).
Pathological TNM after surgery (ypTNM) was compared to ypTNM, and the response was:
-
ypPR in 27 patients (60%).
-
ypCR in 10 patients (22.2%).
-
ypSD in 8 patients (17.7%).
The pCR rates were consistent with literature data supporting the TNT strategy (Table 2) [17].
After surgery, every patient continued trimestral follow-up for the first two years, and semestral follow-up after that. At each visit, patients were clinical and hematological evaluated. Moreover, they performed semestral instrumental evaluation with CT scan (brain, chest, abdomen and pelvis) or colonscopy. To date, after a median follow-up of 30 months, 43 out of 45 patients (95.5%) are still alive, and two patients (4.4%) died of cardiovascular comorbidities without evidence of tumor relapse. Regarding DFS rates, seven patients (15.5%) experienced progression disease (PD); three patients (6.6%) were diagnosed with hepatic metastasis after a median follow-up of 5.5 months; three patients (6.6%) were diagnosed with lung metastasis after a median follow-up of 12 months; and one patient (2.2%) had local relapse (presacral) after 13 months (Figure 3).

4. Discussion

While surgery is considered the main treatment for early-stage rectal cancer, locally advanced rectal cancer needs to be handled with a multidisciplinary approach.
Since the late 1990s, the role of neoadjuvant radiotherapy has been investigated. First, the efficacy of preoperative short-course radiotherapy (SCRT) to reduce rates of local recurrence and improve survival was demonstrated by the Swedish Rectal Cancer Trial and the Dutch Trial. Then, The German CAO/ARO/AIO-94 Trial established preoperative chemoradiotherapy with 5-FU, TME surgery, and postoperative chemotherapy with 5-FU as standard treatment for LARC [4,5,6].
Currently, different evidence-based approaches are supported for LARC treatment. Concurrent CRT and LCRT is generally performed in the United States and several European countries. It delivers 45–50 Gy of external-beam, intensity-modulated radiation therapy over 25 to 28 daily fractions, with concomitant radiation-sensitizing, fluoropyrimidine-based chemotherapy such as oral capecitabine, continuous infusion of 5-FU or bolus 5-FU with leucovorin, followed by delayed surgery 6–8 weeks later. SCRT is preferred in Northern European countries. It aims to sterilize the mesorectal fat before surgery, and the regimen does not include chemotherapy. Patients receive 5 Gy per day for a total of 5 days, then typically proceed to surgery within 1–2 weeks [18,19,20,21].
Surgery is performed according to different techniques, such as sphincter-saving low anterior resection (LAR), or non-sphincter-saving abdomino-perineal resection (APR), based on preoperative staging. Both approaches include TME to ensure margins and lymph node retrieval [22].
In the postoperative setting, additional chemotherapy can be performed according to different schemes, such as: FOLFOX4 (oxaliplatin, 5-FU, and leucovorin) regimen, 5-FU and leucovorin, CAPOX (capecitabine plus oxaliplatin), or capecitabine alone [23]. Even if the use of adjuvant chemotherapy is recommended by different international guidelines, its use in patients who undergo preoperative CRT is controversial. Results of recent multicentre randomised control trials showed no benefit in terms of survival and distant metastases rates. Therefore, its use routinely requires rigorous evaluation [24,25,26].
The administration of this multimodal approach accounted for reduction in the local pelvic recurrence rates to less than 10%, but the possibility of distant metastasis remains more than twice that of primary tumour recurrence; furthermore, it is not free from complications. Literature data suggest that because of poor postoperative physical condition, more than a third of the patients delay or refuse adjuvant chemotherapy, and less than half of the eligible patients receive the full course of chemotherapy [27].
In the interest of improving compliance rates, reducing toxicity, and decreasing distant relapse rates, multiple prospective trials investigated the advantages of shifting systemic therapy delivery earlier in the treatment paradigm, incorporating induction or consolidation chemotherapy and chemoradiotherapy in the neoadjuvant setting, supporting the administration of a novel approach termed total neoadjuvant therapy.
The theoretical considerations which provide a solid rationale for TNT are: the potential to optimally treat preoperative occult micrometastatic disease, the possibility of delivering chemotherapy agents directly to the primary tumour while it has a fully intact vasculature (undisrupted by radiation or surgery), the prospect of reducing the duration of temporary ostomies, and avoiding the challenges of chemotherapy with an ostomy, improving the quality of life after surgery [28,29].
On these bases, the value of this approach has been recently investigated [30,31,32]. In 2020, TNT has been recommended as acceptable for stage II and III LARC by the National Comprehensive Cancer Network (NCCN) [33].
In 2021, the first two large, randomised phase III trials comparing TNT with the standard approach, in both cases CRT, released their preliminary results. The RAPIDO trial compared SCRT, followed by CAPOX or FOLFOX4 consolidation chemotherapy, followed by surgery (without adjuvant treatment) with CRT, followed by surgery and CAPOX or FOLFOX4 adjuvant chemotherapy. The PRODIGE-23 trial compared modified FOLFIRINOX followed by CRT, surgery and capecitabine or modified FOLFOX6 adjuvant chemotherapy with CRT, followed by surgery and capecitabine or modified FOLFOX6 adjuvant chemotherapy [34,35,36].
The primary endpoint was 3-year disease-related treatment failure for the RAPIDO trial and 3-year disease-free survival for PRODIGE 23. Both studies met their respective primary endpoints with statistically significant hazard ratios, paving the way for a new practical approach [37,38,39].
Currently, total neoadjuvant therapy can be performed following three different strategies: chemoradiotherapy followed by consolidation chemotherapy; induction chemotherapy followed by chemoradiotherapy; and induction chemotherapy, followed by chemoradiotherapy, followed by consolidation chemotherapy (sandwich TNT) [40,41].
To date, the most-performed induction and consolidation chemotherapy schemes are capecitabine-based, or 5-FU based. The role of bevacizumab, a monoclonal antibody against vascular endothelial growth factor (VEGF), has been lately investigated; however, due to the small amount of relevant data, further studies are needed [42,43,44].
A systematic review published in 2021 reported that patients receiving induction chemotherapy type TNT are 28% more likely to have complete pathological response, 44% less likely to have residual nodal disease at surgery, and 57% less likely to have positive margins, while patients receiving consolidation chemotherapy type TNT are 90% more likely to have complete pathological response, even if it does not provide a better nodal downstaging, and has similar positive margin rates to standard long course CRT. Sandwich TNT has been demonstrated to be highly effective in terms of pCR and major regression [45,46,47].
Basing on literature data suggesting promising advantages of TNT strategy, we performed a retrospective, single-arm, single-center study on 45 patients affected by histologically and radiologically proven locally advanced rectal cancer, with the aim of analyzing the feasibility and toxicity rates of an integrated intensified treatment in the setting of TNT. Moreover, we investigated short-term TNT efficacy, in terms of pCR rates and DFS.
All patients were candidates to receive three cycles of FOLFOX4 (35 patients) or De Gramont (10 patients) induction chemotherapy, followed by concurrent CRT with long-course radiotherapy plus concomitant boost and continuous infusion of 5-FU (45 patients), followed by three cycles of FOLFOX4 (32 patients) or De Gramont (13 patients) consolidation chemotherapy.
Surgery was performed approximately eight weeks after the end of TNT.
The response obtained after TNT allowed surgeons to perform sphincter-saving surgery in 10 out of 18 patients initially selected for abdomino-perineal resection (distal tumour location < 6 cm).
All the chemoradiotherapy schemes were well tolerated; the majority of patients presented mild adverse events related to oxaliplatin or fluoropyrimidines.
The PCR rates (22.2%) and DFS at a median follow-up of 30 months (80%) are consistent with literature data and support the efficacy of TNT in terms of short-term outcomes. Limitations of this study are that the selected population is relatively poor, and a longer median follow-up is needed to evaluate the real advantages of this strategy.

5. Conclusions

Our treatment protocol, based on integrated intensified treatment in the setting of total neoadjuvant therapy, has shown to be feasible and well tolerated, with only mild adverse events related to oxaliplatin or fluoropyrimidines.
Even if it is currently only possible to evaluate short-term outcomes, the high rate of pCR (22.2%) and DFS at a median follow-up of 30 months (80%) is encouraging. However, long-term follow-up is still required to determine if these advantages translate into improved overall survival.

Author Contributions

Conceptualization, M.C.L.G., R.L.E.L., R.M. and C.S.; Investigation, M.L.R., G.M., I.F., R.L.E.L., R.M., G.A. and S.P. (Silvana Parisi); Methodology, M.L.R., G.M., I.F., G.A. and S.P. (Silvana Parisi); Resources, A.B., S.P. (Stefano Palmucci), P.V.F., S.P. (Stefano Pergolizzi) and A.P.; Writing—original draft, M.C.L.G. and C.S.; Writing—review & editing, S.P. (Stefano Pergolizzi), A.P. and C.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The present study was conducted in accordance with the Declaration of Helsinki, ICH-GCP, and all applicable laws and requirements. The trial received approvals from the institutional ethics committee (A.O.U. Policlinico “G. Rodolico-San Marco). A protocol code was not provided because the study did not require the execution of therapeutic interventions in addition to the standard clinical management. Patient confidentiality was maintained by anonymizing patient data to remove any identifying information.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to privacy restrictions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Baidoun, F.; Elshiwy, K.; Elkeraie, Y.; Merjaneh, Z.; Khoudari, G.; Sarmini, M.T.; Gad, M.; Al-Husseini, M.; Saad, A. Colorectal Cancer Epidemiology: Recent Trends and Impact on Outcomes. Curr. Drug Targets 2021, 22, 998–1009. [Google Scholar] [PubMed]
  2. Enker, W.E. Total mesorectal excision—The new golden standard of surgery for rectal cancer. Ann. Med. 1997, 29, 127–133. [Google Scholar] [CrossRef] [PubMed]
  3. Vignali, A.; Elmore, U.; Milone, M.; Rosati, R. Transanal total mesorectal excision (TaTME): Current status and future perspectives. Updat. Surg. 2019, 71, 29–37. [Google Scholar] [CrossRef] [PubMed]
  4. Cedermark, B.; Dahlberg, M.; Glimelius, B.; Påhlman, L.; Rutqvist, L.E.; Wilking, N. Improved survival with preoperative radiotherapy in resectable rectal cancer. New Engl. J. Med. 1997, 336, 980–987. [Google Scholar] [PubMed]
  5. Kapiteijn, E.; Marijnen, C.A.; Nagtegaal, I.D.; Putter, H.; Steup, W.H.; Wiggers, T.; Rutten, H.J.; Pahlman, L.; Glimelius, B.; van Krieken, J.H.; et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. New Engl. J. Med. 2001, 345, 638–646. [Google Scholar] [CrossRef] [PubMed]
  6. Sauer, R.; Fietkau, R.; Wittekind, C.; Rödel, C.; Martus, P.; Hohenberger, W.; Tschmelitsch, J.; Sabitzer, H.; Karstens, J.H.; Becker, H.; et al. Adjuvant vs. neoadjuvant radiochemotherapy for locally advanced rectal cancer: The German trial CAO/ARO/AIO-94. Color. Dis. Off. J. Assoc. Coloproctol. Great Br. Irel. 2003, 5, 406–415. [Google Scholar] [CrossRef]
  7. Ma, Z.; Tan, L.; Liu, Z.L.; Xiao, J.W. Total neoadjuvant therapy or standard chemoradiotherapy for locally advanced rectal cancer: A systematic review and meta-analysis. Front. Surg. 2022, 9, 911538. [Google Scholar] [CrossRef]
  8. Jessup, M.J.; Goldberg, R.M.; Asare, E.A.; Benson, A.B., III; Brierley, J.D.; Chang, G.J.; Chen, V.; Compton, C.C.; De Nardi, P.; Goodman, K.A.; et al. Colon and rectum. In AJCC Cancer Staging Manual, 8th ed.; Amin, M.B., Greene, F.L., Edge, S.B., Compton, C.C., Gershenwald, J.E., Brookland, R.K., Meyer, L., Gress, D.M., Byrd, D.R., Winchester, D.P., Eds.; Springer: Berlin/Heidelberg, Germany, 2017; pp. 251–274. [Google Scholar]
  9. National Cancer Institute. Common Terminology Criteria for Adverse Events v.3.0 and v.4.0 (CTCAE). 2011. Available online: http://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.html (accessed on 14 June 2010).
  10. Ontario Health (Quality). DPYD Genotyping in Patients Who Have Planned Cancer Treatment with Fluoropyrimidines: A Health Technology Assessment. Ont. Health Technol. Assess. Ser. 2021, 21, 1–186. [Google Scholar]
  11. Caravatta, L.; Lupattelli, M.; Mantello, G.; Gambacorta, M.A.; Chiloiro, G.; Di Tommaso, M.; Rosa, C.; Gasparini, L.; Morganti, A.G.; Picardi, V.; et al. Treatment Volume, Dose Prescription and Delivery Techniques for Dose-intensification in Rectal Cancer: A National Survey. Anticancer Res. 2021, 41, 1985–1995. [Google Scholar] [CrossRef]
  12. Pointreau, Y.; Moreau, J.; Vendrely, V.; Schipman, B. Quel apport de la modulation d’intensité pour la radiothérapie des cancers du rectum? [Impact of IMRT for neoadjuvant rectal cancer?]. Cancer Radiother. J. Soc. Fr. Radiother. Oncol. 2022, 26, 865–870. [Google Scholar]
  13. Suwinski, R.; Wzietek, I.; Tarnawski, R.; Namysl-Kaletka, A.; Kryj, M.; Chmielarz, A.; Wydmanski, J. Moderately low alpha/beta ratio for rectal cancer may best explain the outcome of three fractionation schedules of preoperative radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2007, 69, 793–799. [Google Scholar] [CrossRef] [PubMed]
  14. Schwartz, L.H.; Seymour, L.; Litière, S.; Ford, R.; Gwyther, S.; Mandrekar, S.; Shankar, L.; Bogaerts, J.; Chen, A.; Dancey, J.; et al. RECIST 1.1—Standardisation and disease-specific adaptations: Perspectives from the RECIST Working Group. Eur. J. Cancer 2016, 62, 138–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Palmucci, S.; Piccoli, M.; Piana, S.; Foti, P.V.; Siverino, R.; Mauro, L.A.; Milone, P.; Ettorre, G.C. Diffusion MRI for rectal cancer staging: ADC measurements before and after ultrasonographic gel lumen distension. Eur. J. Radiol. 2017, 86, 119–126. [Google Scholar] [CrossRef]
  16. Foti, P.V.; Privitera, G.; Piana, S.; Palmucci, S.; Spatola, C.; Bevilacqua, R.; Raffaele, L.; Salamone, V.; Caltabiano, R.; Magro, G.; et al. Locally advanced rectal cancer: Qualitative and quantitative evaluation of diffusion-weighted MR imaging in the response assessment after neoadjuvant chemo-radiotherapy. Eur. J. Radiol. Open 2016, 3, 145–152. [Google Scholar] [CrossRef]
  17. Kasi, A.; Abbasi, S.; Handa, S.; Al-Rajabi, R.; Saeed, A.; Baranda, J.; Sun, W. Total Neoadjuvant Therapy vs. Standard Therapy in Locally Advanced Rectal Cancer: A Systematic Review and Meta-analysis. JAMA Netw. Open 2020, 3, 12. [Google Scholar] [CrossRef]
  18. Spatola, C.; Privitera, G.; Milazzotto, R.; Tocco, A.; Acquaviva, G.; Marletta, F.; Marino, L.; Di Grazia, A.; Salvo, R.; Cartia, G.; et al. Trends in combined radio-chemotherapy for locally advanced rectal cancer: A survey among radiation oncology centers of Sicily region on behalf of AIRO. Radiol. Med. 2019, 124, 671–681. [Google Scholar] [CrossRef]
  19. Spatola, C.; Raffaele, L.; Tocco, A.; Acquaviva, G.; Milazzotto, R.; Bevilacqua, R.; Salamone, V.; Foti, P.V.; Milone, P.; Basile, A.; et al. Intensified neoadjuvant radio-chemotherapy for locally advanced rectal cancer: Mono-istitutional experience and long-term results. Int. J. Radiat. Res. 2019, 17, 265–273. [Google Scholar]
  20. Wang, J.; Long, Y.; Liu, K.; Pei, Q.; Zhu, H. Comparing neoadjuvant long-course chemoradiotherapy with short-course radiotherapy in rectal cancer. BMC Gastroenterol. 2021, 21, 277. [Google Scholar] [CrossRef]
  21. Palta, M.; Willett, C.G.; Czito, B.G. Short-course versus long-course chemoradiation in rectal cancer--time to change strategies? Curr. Treat. Options Oncol. 2014, 15, 421–428. [Google Scholar] [CrossRef]
  22. Delibegovic, S. Introduction to Total Mesorectal Excision. Med. Arch. 2017, 71, 434–438. [Google Scholar] [CrossRef]
  23. Rödel, C.; Graeven, U.; Fietkau, R.; Hohenberger, W.; Hothorn, T.; Arnold, D.; Hofheinz, R.D.; Ghadimi, M.; Wolff, H.A.; Lang-Welzenbach, M.; et al. Oxaliplatin added to fluorouracil-based preoperative chemoradiotherapy and postoperative chemotherapy of locally advanced rectal cancer (the German CAO/ARO/AIO-04 study): Final results of the multicentre, open-label, randomised, phase 3 trial. Lancet. Oncol. 2015, 16, 979–989. [Google Scholar] [CrossRef] [PubMed]
  24. Bregni, G.; Akin Telli, T.; Camera, S.; Deleporte, A.; Moretti, L.; Bali, A.M.; Liberale, G.; Holbrechts, S.; Hendlisz, A.; Sclafani, F. Adjuvant chemotherapy for rectal cancer: Current evidence and recommendations for clinical practice. Cancer Treat. Rev. 2020, 83, 101948. [Google Scholar] [CrossRef] [PubMed]
  25. Bosset, J.F.; Calais, G.; Mineur, L.; Maingon, P.; Stojanovic-Rundic, S.; Bensadoun, R.J.; Bardet, E.; Beny, A.; Ollier, J.C.; Bolla, M.; et al. Fluorouracil-based adjuvant chemotherapy after preoperative chemoradiotherapy in rectal cancer: Long-term results of the EORTC 22921 randomised study. Lancet. Oncol. 2014, 15, 184–190. [Google Scholar] [CrossRef] [PubMed]
  26. Spiegel, D.Y.; Boyer, M.J.; Hong, J.C.; Williams, C.D.; Kelley, M.J.; Salama, J.K.; Palta, M. Survival Advantage with Adjuvant Chemotherapy for Locoregionally Advanced Rectal Cancer: A Veterans Health Administration Analysis. J. Natl. Compr. Cancer Netw. JNCCN 2020, 18, 52–58. [Google Scholar] [CrossRef]
  27. Zhao, Y.; Zhu, J.; Yang, B.; Gao, Q.; Xu, Y.; Wei, X.; Kong, D.; Ji, S.; Fei, B. Retrospective study of total neoadjuvant therapy for locally advanced rectal cancer. Future Oncol. 2022, 18, 691–700. [Google Scholar] [CrossRef]
  28. Deschner, B.W.; VanderWalde, N.A.; Grothey, A.; Shibata, D. Evolution and Current Status of the Multidisciplinary Management of Locally Advanced Rectal Cancer. JCO Oncol. Pract. 2021, 17, 383–402. [Google Scholar] [CrossRef]
  29. Hunt, S.R. Total Neoadjuvant Therapy for Rectal Cancer. In Seminars in Colon & Rectal Surgery; WB Saunders: London, UK, 2019. [Google Scholar]
  30. Negri, F.; Aschele, C. Unconsolidated Results of Consolidation Chemotherapy Following Short-Course Radiotherapy in Locally Advanced Rectal Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 4028. [Google Scholar] [CrossRef]
  31. Jin, J.; Tang, Y.; Hu, C.; Jiang, L.-M.; Jiang, J.; Li, N.; Liu, W.-Y.; Chen, S.-L.; Li, S.; Lu, N.-N.; et al. Multicenter, randomized, phase III trial of short-term radiotherapy plus chemotherapy versus long-term chemoradiotherapy in locally advanced rectal cancer (STELLAR). J. Clin. Oncol. 2022, 40, 1681–1692. [Google Scholar] [CrossRef]
  32. Guida, A.M.; Sensi, B.; Formica, V.; D’Angelillo, R.M.; Roselli, M.; Del Vecchio Blanco, G.; Rossi, P.; Capolupo, G.T.; Caricato, M.; Sica, G.S. Total neoadjuvant therapy for the treatment of locally advanced rectal cancer: A systematic minireview. Biol. Direct 2022, 17, 16. [Google Scholar] [CrossRef]
  33. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Arain, M.A.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Garrido-Laguna, I.; et al. NCCN Guidelines Insights: Rectal Cancer, Version 6. 2020. J. Natl. Compr. Canc. Netw. 2020, 18, 806–815. [Google Scholar] [CrossRef]
  34. Conroy, T.; Bosset, J.F.; Etienne, P.L.; Rio, E.; François, É.; Mesgouez-Nebout, N.; Vendrely, V.; Artignan, X.; Bouché, O.; Gargot, D.; et al. Neoadjuvant chemotherapy with FOLFIRINOX and preoperative chemoradiotherapy for patients with locally advanced rectal cancer (UNICANCER-PRODIGE 23): A multicentre, randomised, open-label, phase 3 trial. Lancet. Oncol. 2021, 22, 702–715. [Google Scholar] [CrossRef] [PubMed]
  35. Bahadoer, R.R.; Dijkstra, E.A.; Van Etten, B.; Marijnen, C.; Putter, H.; Kranenbarg, E.M.; Roodvoets, A.; Nagtegaal, I.D.; Beets-Tan, R.; Blomqvist, L.K.; et al. Short-course radiotherapy followed by chemotherapy before total mesorectal excision (TME) versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer (RAPIDO): A randomised, open-label, phase 3 trial. Lancet. Oncol. 2021, 22, 29–42. [Google Scholar] [CrossRef] [PubMed]
  36. Giunta, E.F.; Bregni, G.; Pretta, A.; Deleporte, A.; Liberale, G.; Bali, A.M.; Moretti, L.; Troiani, T.; Ciardiello, F.; Hendlisz, A.; et al. Total neoadjuvant therapy for rectal cancer: Making sense of the results from the RAPIDO and PRODIGE 23 trials. Cancer Treat. Rev. 2021, 96, 102177. [Google Scholar] [CrossRef] [PubMed]
  37. Papaccio, F.; Roselló, S.; Huerta, M.; Gambardella, V.; Tarazona, N.; Fleitas, T.; Roda, D.; Cervantes, A. Neoadjuvant Chemotherapy in Locally Advanced Rectal Cancer. Cancers 2020, 12, 3611. [Google Scholar] [CrossRef] [PubMed]
  38. Nagarajan, A. Total neoadjuvant therapy: Fact, fantasy, or fallacy? Surg. Oncol. 2022, 43, 101738. [Google Scholar] [CrossRef]
  39. Bhudia, J.; Glynne-Jones, R. The Evolving Neoadjuvant Treatment Paradigm for Patients with Locoregional mismatch Repair Proficient Rectal Cancer. Curr. Treat. Options Oncol. 2022, 23, 453–473. [Google Scholar] [CrossRef]
  40. Hu, Y.H.; Wei, J.W.; Chang, H.; Xiao, W.W.; Lin, J.Z.; Cai, M.Y.; Cai, P.Q.; Kong, L.H.; Chen, G.; Pan, Z.Z.; et al. The high pCR rate of sandwich neoadjuvant treatment in locally advanced rectal cancer may translate into a better long-term survival benefit: 5-year outcome of a Phase II clinical trial. Cancer Manag. Res. 2018, 10, 4363–4369. [Google Scholar] [CrossRef]
  41. Petrelli, F.; Trevisan, F.; Cabiddu, M.; Sgroi, G.; Bruschieri, L.; Rausa, E.; Ghidini, M.; Turati, L. Total Neoadjuvant Therapy in Rectal Cancer: A Systematic Review and Meta-analysis of Treatment Outcomes. Ann. Surg. 2020, 271, 440–448. [Google Scholar] [CrossRef]
  42. Tuta, M.; Boc, N.; Brecelj, E.; Omejc, M.; Anderluh, F.; Ermenc, A.S.; Peressutti, A.J.; Oblak, I.; Krebs, B.; Velenik, V. Total neoadjuvant treatment of locally advanced rectal cancer with high risk factors in Slovenia. Radiol. Oncol. 2019, 53, 465–472. [Google Scholar] [CrossRef]
  43. Zhou, Y.; Guo, Z.; Wu, Z.; Shi, J.; Zhou, C.; Sun, J.; Hidasa, I.; Lu, X.; Lu, C. The efficacy and safety of adding bevacizumab in neoadjuvant therapy for locally advanced rectal cancer patients: A systematic review and meta-analysis. Transl. Oncol. 2021, 14, 100964. [Google Scholar] [CrossRef]
  44. Masi, G.; Vivaldi, C.; Fornaro, L.; Lonardi, S.; Buccianti, P.; Sainato, A.; Marcucci, L.; Martignetti, A.; Luca Urso, E.D.; Castagna, M.; et al. Total neoadjuvant approach with FOLFOXIRI plus bevacizumab followed by chemoradiotherapy plus bevacizumab in locally advanced rectal cancer: The TRUST trial. Eur. J. Cancer 2019, 110, 32–41. [Google Scholar] [CrossRef] [PubMed]
  45. Kong, J.C.; Soucisse, M.; Michael, M.; Tie, J.; Ngan, S.Y.; Leong, T.; McCormick, J.; Warrier, S.K.; Heriot, A.G. Total Neoadjuvant Therapy in Locally Advanced Rectal Cancer: A Systematic Review and Metaanalysis of Oncological and Operative Outcomes. Ann. Surg. Oncol. 2021, 28, 7476–7486. [Google Scholar] [CrossRef] [PubMed]
  46. Feng, W.; Yu, B.; Zhang, Z.; Li, J.; Wang, Y. Current status of total neoadjuvant therapy for locally advanced rectal cancer. Asia-Pac. J. Clin. Oncol. 2021, 18, 546–559. [Google Scholar] [CrossRef] [PubMed]
  47. Gao, Y.H.; Lin, J.Z.; An, X.; Luo, J.L.; Cai, M.Y.; Cai, P.Q.; Kong, L.H.; Liu, G.C.; Tang, J.H.; Chen, G.; et al. Neoadjuvant sandwich treatment with oxaliplatin and capecitabine administered prior to, concurrently with, and following radiation therapy in locally advanced rectal cancer: A prospective phase 2 trial. Int. J. Radiat. Oncol. Biol. Phys. 2014, 90, 1153–1160. [Google Scholar] [CrossRef]
Figure 1. Consort diagram showing the different application and feasibility of preoperative induction chemotherapy (FOLFOX4, De Gramont) radio-chemotherapy (CFRT, HFRT), and consolidation chemotherapy (FOLFOX4, De Gramont).
Figure 1. Consort diagram showing the different application and feasibility of preoperative induction chemotherapy (FOLFOX4, De Gramont) radio-chemotherapy (CFRT, HFRT), and consolidation chemotherapy (FOLFOX4, De Gramont).
Cancers 15 00921 g001
Figure 2. Comparation between pathological and radiological response.
Figure 2. Comparation between pathological and radiological response.
Cancers 15 00921 g002
Figure 3. Kaplan–Meier curves showing DFS rates comparing different risk groups (cTNM, patological response after neoadjuvant therapy).
Figure 3. Kaplan–Meier curves showing DFS rates comparing different risk groups (cTNM, patological response after neoadjuvant therapy).
Cancers 15 00921 g003
Table 1. Patients’ characteristics.
Table 1. Patients’ characteristics.
CharacteristicsN. of PatientsPercentage
SexMales3066.6%
Females1533.3%
Age<54 y.o.715.5%
55–64 y.o.1226.6%
65–74 y.o.1022.2%
>75 y.o.1635.5%
KPS100%2760%
90%1431.1%
80%48.8%
ComorbiditiesNone1124.4%
Diabetes1124.4%
Hypertension2657.7%
Hypercholesterolemia613.3%
Onset symptomsRectorrhagia2044.4%
Abdominal pain2044.4%
Irregular alvus1840%
Tenesmus1737.7%
Substruction/obstruction1226.6%
GradingG11022.2%
G21533.3%
G32044.4%
Distance from mesorectal fascia>1 mm3168.8%
<1 mm1431.1%
Distance from A.O.<6 cm1840%
6.1–10 cm2248.8%
>10 cm511.1%
StageT2 N1-2715.5%
T3-T4 N0817.7%
T3-T4 N1-23066.6%
Table 2. Literature data supporting the TNT strategy [17].
Table 2. Literature data supporting the TNT strategy [17].
SourceNo. of Events/TotalPercentage
Calvo et al. 200615/5228.8%
Cerek et al. 2018110/30835.7%
Conroy et al. 202064/23127.7%
Garcia-Aguilar et al. 201525/6538.4%
Markovina et al. 201719/6927.5%
Van der Valk et al. 2020117/42327.6%
Van Zoggel et al. 201810/5817.24%
Our Study10/4522.2%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lo Greco, M.C.; La Rocca, M.; Marano, G.; Finocchiaro, I.; Liardo, R.L.E.; Milazzotto, R.; Acquaviva, G.; Basile, A.; Palmucci, S.; Foti, P.V.; et al. Integrated Intensified Chemoradiation in the Setting of Total Neoadjuvant Therapy (TNT) in Patients with Locally Advanced Rectal Cancer: A Retrospective Single-Arm Study on Feasibility and Efficacy. Cancers 2023, 15, 921. https://doi.org/10.3390/cancers15030921

AMA Style

Lo Greco MC, La Rocca M, Marano G, Finocchiaro I, Liardo RLE, Milazzotto R, Acquaviva G, Basile A, Palmucci S, Foti PV, et al. Integrated Intensified Chemoradiation in the Setting of Total Neoadjuvant Therapy (TNT) in Patients with Locally Advanced Rectal Cancer: A Retrospective Single-Arm Study on Feasibility and Efficacy. Cancers. 2023; 15(3):921. https://doi.org/10.3390/cancers15030921

Chicago/Turabian Style

Lo Greco, Maria Chiara, Madalina La Rocca, Giorgia Marano, Irene Finocchiaro, Rocco Luca Emanuele Liardo, Roberto Milazzotto, Grazia Acquaviva, Antonello Basile, Stefano Palmucci, Pietro Valerio Foti, and et al. 2023. "Integrated Intensified Chemoradiation in the Setting of Total Neoadjuvant Therapy (TNT) in Patients with Locally Advanced Rectal Cancer: A Retrospective Single-Arm Study on Feasibility and Efficacy" Cancers 15, no. 3: 921. https://doi.org/10.3390/cancers15030921

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop