ArticlePDF Available

Taxane-Based Chemotherapy Is Effective in Metastatic Appendiceal Adenocarcinoma

Authors:

Abstract and Figures

Appendiceal cancer is a rare, orphan disease with no therapies currently approved by the FDA for its treatment. Given the limited data regarding drug efficacy, these tumors have historically been treated with chemotherapy designed for colon cancer. However, an overwhelming body of molecular data has demonstrated that appendiceal adenocarcinoma is a distinct entity with key molecular differences from colon cancer, notably rare APC mutation. Recognizing that APC loss-of-function is thought to contribute to taxane resistance and that taxanes are effective in the treatment of other gastrointestinal tumors, including gastric, esophageal, and small bowel adenocarcinoma, we completed a single-center retrospective study to assess efficacy. In a cohort of 13 patients with metastatic appendiceal adenocarcinoma, treated with taxane chemotherapy the median overall survival was 8.8 months. Of 10 evaluable patients, we observed 3 responses, 4 patients with stable disease, and 3 with progression (30% response rate, 70% disease control rate). The results of this study showing activity of taxane-based chemotherapy in appendiceal adenocarcinoma support further clinical investigation of taxane therapy in this orphan disease.
Content may be subject to copyright.
The Oncologist, 2023, 28, e1303–e1305
https://doi.org/10.1093/oncolo/oyad263
Advance access publication 20 September 2023
Brief Communication
© The Author(s) 2023. Published by Oxford University Press.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/
licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For
commercial re-use, please contact journals.permissions@oup.com.
Received: 29 June 2023; Accepted: 24 August 2023.
Taxane-Based Chemotherapy Is Effective in Metastatic
Appendiceal Adenocarcinoma
JuliaDansby1, AdityaMore1, MohammadZeineddine1, AbdelrahmanYousef1, AlishaBent1,
FarshidDayyani2, RobertWolff1,, MichaelOverman1,, John PaulShen*,1,
1Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
2Division of Hematology/Oncology, UC Irvine Health, Irvine, CA, USA
*Corresponding author: John Paul Shen, MD, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515
Holcombe Blvd., Houston, TX 77030, USA. Email: jshen8@mdanderson.org
Abstract
Appendiceal cancer is a rare, orphan disease with no therapies currently approved by the FDA for its treatment. Given the limited data regarding
drug efficacy, these tumors have historically been treated with chemotherapy designed for colon cancer. However, an overwhelming body of
molecular data has demonstrated that appendiceal adenocarcinoma is a distinct entity with key molecular differences from colon cancer, notably
rare APC mutation. Recognizing that APC loss-of-function is thought to contribute to taxane resistance and that taxanes are effective in the
treatment of other gastrointestinal tumors, including gastric, esophageal, and small bowel adenocarcinoma, we completed a single-center ret-
rospective study to assess efficacy. In a cohort of 13 patients with metastatic appendiceal adenocarcinoma, treated with taxane chemotherapy
the median overall survival was 8.8 months. Of 10 evaluable patients, we observed 3 responses, 4 patients with stable disease, and 3 with pro-
gression (30% response rate, 70% disease control rate). The results of this study showing activity of taxane-based chemotherapy in appendiceal
adenocarcinoma support further clinical investigation of taxane therapy in this orphan disease.
Key words: appendiceal cancer; taxane chemotherapy.
Introduction
Appendiceal tumors encompass a rare and diverse group of
neoplasms, with appendiceal adenocarcinoma (AA) being
the most common histologic subtype.1 Despite a unique nat-
ural history characterized by metastatic spread limited to
the peritoneum, as well as growing evidence that appendi-
ceal tumors are molecularly distinct from colorectal cancer
(CRC),2 current National Comprehensive Cancer Network
(NCCN) guidelines suggest that appendiceal tumors should
be treated with the same chemotherapy as used for CRC.
However, recent prospective and retrospective studies have
called into question the dogma that appendiceal tumors re-
spond to chemotherapy similarly to colon cancer.3,4 In par-
ticular, a prospective, randomized, crossover design trial
showed that patients with low-grade mucinous AAs do not
derive benet from 5-FU-based chemotherapy,4 highlighting
the need to drug development efforts specic to appendix
cancer.
Taxanes have been shown to be ineffective in colorectal
cancer but are active in small bowel adenocarcinoma (SBA).5
It has been reported that APC loss-of-function is a mecha-
nism of taxane resistance.6 Unlike CRC, where APC is mu-
tated in >70% of tumors, APC mutation is uncommon in
subtypes of appendiceal cancer (9.0%).2 We, therefore, hy-
pothesized that taxane chemotherapy would have activity in
AA.
Materials and Methods
The MD Anderson adapted version of the Palantir-Foundry
software system was used to perform an automated query of
the MD Anderson GI Medical Oncology database to identify
patients with AA treated with paclitaxel-based regimens be-
tween 2003 and 2022. Manual chart review was performed to
conrm patients met eligibility criteria and to extract outcome
variables. Eligible patients had pathologic diagnosis of AA,
mucinous adenocarcinoma, signet ring cell adenocarcinoma,
or goblet cell adenocarcinoma, and more than one dose of
taxane-based therapy and were not enrolled in a clinical tri-
al. Radiographic response was assessed retrospectively based
upon the treating physician’s assessment and categorized as
response, stable disease, progression; response could not be
evaluated (NA) if the patient did not have imaging performed
after starting taxane therapy. Biochemical response was eval-
uated based on the percent change in tumor marker before
and after treatment. Patients with a greater than 20% decrease
were categorized as response, less than 20% change as stable,
and greater than 20% increase as progression. Those whose
respective tumor markers were not elevated were indicated as
such, and those whose tumor markers were not measured were
classied as NA. In cases where biochemical response differed
from radiographic response, radiographic response was used
for nal response determination. Median overall survival (OS)
was determined using the Kaplan-Meier method.
Downloaded from https://academic.oup.com/oncolo/article/28/12/e1303/7279402 by guest on 13 December 2023
e1304 The Oncologist, 2023, Vol. 28, No. 12
Results
Thirteen patients with AA treated with paclitaxel-based
therapy were identied and met inclusion criteria. Median
age was 64 years (range 29-77) with roughly equal splits
between male and female as well as well, moderate, and
poorly differentiated tumors (Supplementary Table S1).
All 13 patients had inoperable peritoneal disease at time
of treatment, and 4 (30.1%) had prior surgical resection.
The cohort was heavily pretreated with median of 3 prior
lines of therapy (range 1-5), 10 of the 13 had prior treat-
ment with either FOLFOX or CapeOx. Ten patients received
taxane-based combinations (gemcitabine combination in 7,
platinum combination in 2, and uoropyrimidine combina-
tion in 1), 3 received paclitaxel monotherapy. Four of the
combination-treated patients received nab-paclitaxel, the
rest paclitaxel.
The median OS from the start of taxane therapy was 8.77
months (range 0.7-31.0 months, Fig. 1A) with 3 of 13 pa-
tients still alive at time of analysis. Four patients stopped
therapy after 3 or fewer taxane treatments due to either
small bowel obstruction (SBO) or deteriorating perfor-
mance status, median progression-free survival (PFS) for
the remaining 9 patients was 7.4 months (range 0.8-15.5
months, Supplementary Table S1). Median time on treat-
ment was 2.5 months (range 0.2-13.1 months, Fig. 1B)
with 3 patients remaining on treatment at time of analysis.
Of the 10 patients that could be assessed for radiographic
response, 3 showed response, 4 with stable disease, and 3
with progression for a response rate of 30% and a disease
control rate of 70% (Fig. 1C). Of the 9 patients with elevat-
ed CEA, biochemical response was seen in 3 (33%), stable
disease in 3 (33%), and progression in 3 (33%). Of the 6
patients with elevated CA 19-9, biochemical response was
seen in 2 (33%), stable disease in 2 (33%), and progres-
sion in 2 (33%). Two patients received rst-line carbopla-
tin and paclitaxel, both had response; the third responding
patient was treated with gemcitabine and nab-paclitaxel.
Presumably, the addition of a second cytotoxic agent con-
tributed to response, however, the activity of carboplatin
and gemcitabine in appendiceal cancer is unknown. One pa-
tient was initially diagnosed with a primary ovarian tumor
and changed to 5-FU and bevacizumab after 3 cycles once
expert pathology review made diagnosis of AA. The second
patient had concurrent diagnoses of stage III squamous cell
carcinoma of the lung and goblet cell adenocarcinoma of
the appendix and was treated initially with weekly carbo-
platin and paclitaxel with concurrent radiation and then
carboplatin and paclitaxel plus pembrolizumab per NSCLC
guidelines. Treatment with carboplatin and paclitaxel is on-
going, with marked radiographic improvement in peritone-
al carcinomatosis and complete response of NSCLC based
on imaging and endobronchial biopsies. There was not an
associated between grade and radiographic response. Three
of four GNAS mutant tumors did not respond, consistent
with prior reports suggesting intrinsic resistance to therapy
in the case of mutant GNAS.3
Discussion
Complete cytoreductive surgery (CRS) with heated intra-
peritoneal chemotherapy (HIPEC) remains the treatment
of choice for patients with metastatic AA;7 however, for
many patients the extent of peritoneal metastatic disease
precludes this treatment option. There is unfortunately
very little prospective data to guide chemotherapy choice
for these nonoperative patients. To our knowledge, this
is the largest cohort of patients with appendiceal cancer
treated with taxane-based therapy reported in the liter-
ature. Although we recognize the inherent limitations of
this small, retrospective study design, the favorable disease
control rate in a heavily pretreated cohort indicates that
taxane-based chemotherapy is active in AA and should be
further studied in a prospective fashion. These ndings are
consistent with the activity of taxanes in SBA5,8 and are
also consistent with the activity of intraperitoneal injec-
tion of paclitaxel in orthotopic PDX models of appendiceal
cancer.9,10
Funding
This work was supported by the Col. Daniel Connelly
Memorial Fund, the National Cancer Institute
(K22 CA234406) to J.P.S., and the Cancer Center Support
Grant (P30 CA016672), the Cancer Prevention & Research
Institute of Texas (RR180035 to J.P.S., a CPRIT Scholar in
Cancer Research), and a Conquer Cancer Career Development
Award (CDA-7604125121 to J.P.S). to J.P.S. Any opinions,
ndings, and conclusions expressed in this material are those
Figure 1. Outcomes for patients with AA treated with taxane-based therapy. (A) Overall survival from the start of treatment. (B) Time on treatment. (C)
Radiographic response.
Downloaded from https://academic.oup.com/oncolo/article/28/12/e1303/7279402 by guest on 13 December 2023
e1305The Oncologist, 2023, Vol. 28, No. 12
of the author(s) and do not necessarily reect those of the
American Society of Clinical Oncology or Conquer Cancer.
Conflict of Interests
Farshid Dayyani reported consulting/advisory relationships
Eisai, Exelixis, Ipsen, and AstraZeneca; honoraria from
Sirtex, Ipsen, and Servier; and research funding to institution
from Amgen, AstraZeneca, BMS, Eisai, Exelixis, Roche, Ipsen,
Natera, and Taiho. Robert Wolff reported royalty payments
from McGraw-Hill, Coeditor of MD Anderson Manual of
Medical Oncology. Michael Overman serves on the scien-
tic advisory board for ACPMP. John Paul Shen reported
consulting/advisory relationships with Engine Biosciences
and NaDeNo Nanoscience; research funding from the NIH,
ASCO, and CPRIT; medical advisory board for ACPMP. The
other authors indicated no nancial relationships.
Author Contributions
Conception/design: M.O., J.P.S. Provision of study material
or patients: A.B., M.O., F.D., R.W., J.P.S Collection and/or as-
sembly of data: J.D., A.M., M.Z., A.Y., J.P.S. Data analysis
and interpretation: J.D., J.P.S. Manuscript writing: J.D., J.P.S.
Final approval of manuscript: All authors.
Data Availability
The data underlying this article are available in the article and
in its online supplementary material.
Supplementary Material
Supplementary material is available at The Oncologist online.
References
1. Raghav K, Shen JP, Jácome AA, et al. Integrated clinico-molecular pro-
ling of appendiceal adenocarcinoma reveals a unique grade-driven
entity distinct from colorectal cancer. Br J Cancer. 2020;123(8):1262-
1270. https://doi.org/10.1038/s41416-020-1015-3
2. Ang CS-P, Shen JP, Hardy-Abeloos CJ, et al. Genomic landscape of
appendiceal neoplasms. JCO Precis Oncol. 2018;2(2):1-18. https://
doi.org/10.1200/po.17.00302
3. Foote MB, Walch H, Chatila W, et al. Molecular classication of
appendiceal adenocarcinoma. J Clin Oncol. 2022;41(8):1553-
1564. https://doi.org/10.1200/jco.22.01392
4. Shen JP, Yousef AM, Zeineddine FA, et al. Efcacy of systemic che-
motherapy in patients with low-grade mucinous appendiceal ade-
nocarcinoma: a randomized crossover trial. JAMA Network Open.
2023;6(6):e2316161-e2316161. https://doi.org/10.1001/jamanet-
workopen.2023.16161
5. Overman MJ, Adam L, Raghav K, et al. Phase II study of nab-
paclitaxel in refractory small bowel adenocarcinoma and CpG is-
land methylator phenotype (CIMP)-high colorectal cancer. Ann
Oncol. 2018;29(1):139-144. https://doi.org/10.1093/annonc/mdx688
6. Astarita EM, Maloney SM, Hoover CA, et al. Adenomatous pol-
yposis coli loss controls cell cycle regulators and response to pacli-
taxel in MDA-MB-157 metaplastic breast cancer cells. PLoS One.
2021;16(8):e0255738. https://doi.org/10.1371/journal.pone.0255738
7. Chicago Consensus Working Group. The Chicago Consensus on
peritoneal surface malignancies: management of appendiceal neo-
plasms. Cancer. 2020;126(11):2525-2533.
8. Aldrich JD, Raghav KPS, Varadhachary GR, Wolff RA, Overman
MJ. Retrospective analysis of taxane-based therapy in small bowel
adenocarcinoma. Oncologist. 2019;24(6):e384-e386. https://doi.
org/10.1634/theoncologist.2018-0573
9. Sugarbaker PH. Intraperitoneal paclitaxel: pharmacology, clinical
results and future prospects. J Gastrointest Oncol. 2021;12(Suppl
1):S231-S239. https://doi.org/10.21037/jgo-2020-03
10. Ito I, Yousef AMG, Chowdhury S, et al. Intraperitoneal paclitaxel
is a safe and effective therapeutic strategy for treating mucinous
appendiceal adenocarcinoma. Cancer Res. 2023.
Downloaded from https://academic.oup.com/oncolo/article/28/12/e1303/7279402 by guest on 13 December 2023
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Importance: Appendiceal adenocarcinoma is a rare tumor, and given the inherent difficulties in performing prospective trials in such a rare disease, there are currently minimal high-quality data to guide treatment decisions, highlighting the need for more preclinical and clinical investigation for this disease. Objective: To prospectively evaluate the effectiveness of fluoropyrimidine-based systemic chemotherapy in patients with inoperable low-grade mucinous appendiceal adenocarcinoma. Design, setting, and participants: This open-label randomized crossover trial recruited patients at a single tertiary care comprehensive cancer center from September 2013 to January 2021. The data collection cutoff was May 2022. Enrollment of up to 30 patients was planned. Eligible patients had histological evidence of a metastatic low-grade mucinous appendiceal adenocarcinoma, with radiographic imaging demonstrating the presence of mucinous peritoneal carcinomatosis and were not considered candidates for complete cytoreductive surgery. Key exclusion criteria were concurrent or recent investigational therapy, evidence of bowel obstruction, and use of total parenteral nutrition. Data were analyzed from November 2021 to May 2022. Interventions: Patients were randomized to either 6 months observation followed by 6 months of chemotherapy, or initial chemotherapy followed by observation. Main outcomes and measures: The primary end point was the percentage difference in tumor growth in treatment and observation groups. Key secondary end points included patient-reported outcomes in the chemotherapy and observation periods, objective response rate, rate of bowel complications, and differences in overall survival (OS). Results: A total of 24 patients were enrolled, with median (range) age of 63 (38 to 82) years, and equal proportion of men and women (eg, 12 men [50%]); all patients had ECOG performance status of 0 or 1. A total of 11 patients were randomized to receive chemotherapy first, and 13 patients were randomized to receive observation first. Most patients (15 patients [63%]) were treated with either fluorouracil or capecitabine as single agent; 3 patients (13%) received doublet chemotherapy (leucovorin calcium [folinic acid], fluorouracil, and oxaliplatin or folinic acid, fluorouracil, and irinotecan hydrochloride), and bevacizumab was added to cytotoxic chemotherapy for 5 patients (21%). Fifteen patients were available to evaluate the primary end point of difference in tumor growth during treatment and observation periods. Tumor growth while receiving chemotherapy increased 8.4% (95% CI, 1.5% to 15.3%) from baseline but was not significantly different than tumor growth during observation (4.0%; 95% CI, -0.1% to 8.0%; P = .26). Of 18 patients who received any chemotherapy, none had an objective response (14 patients [77.8%] had stable disease; 4 patients [22.2%] had progressive disease). Median (range) OS was 53.2 (8.1 to 95.5) months, and there was no significant difference in OS between the observation-first group (76.0 [8.6 to 95.5] months) and the treatment-first group (53.2 [8.1 to 64.1] months; hazard ratio, 0.64; 95% CI, 0.16-2.55; P = .48). Patient-reported quality-of-life metrics identified that during treatment, patients experienced significantly worse fatigue (mean [SD] score, 18.5 [18.6] vs 28.9 [21.3]; P = .02), peripheral neuropathy (mean [SD] score, 6.67 [12.28] vs 38.89 [34.88]; P = .01), and financial difficulty (mean [SD] score, 8.9 [15.2] vs 28.9 [33.0]; P = .001) compared with during observation. Conclusions and relevance: In this prospective randomized crossover trial of systemic chemotherapy in patients with low-grade mucinous appendiceal adenocarcinoma, patients did not derive clinical benefit from fluorouracil-based chemotherapy, given there were no objective responses, no difference in OS when treatment was delayed 6 months, and no difference in the rate of tumor growth while receiving chemotherapy. Trial registration: ClinicalTrials.gov Identifier: NCT01946854.
Article
Full-text available
Adenomatous Polyposis Coli (APC) is lost in approximately 70% of sporadic breast cancers, with an inclination towards triple negative breast cancer (TNBC). TNBC is treated with traditional chemotherapy, such as paclitaxel (PTX); however, tumors often develop drug resistance. We previously created APC knockdown cells (APC shRNA1) using the human TNBC cells, MDA-MB-157, and showed that APC loss induces PTX resistance. To understand the mechanisms behind APC-mediated PTX response, we performed cell cycle analysis and analyzed cell cycle related proteins. Cell cycle analysis indicated increased G2/M population in both PTX-treated APC shRNA1 and parental cells, suggesting that APC expression does not alter PTX-induced G2/M arrest. We further studied the subcellular localization of the G2/M transition proteins, cyclin B1 and CDK1. The APC shRNA1 cells had increased CDK1, which was preferentially localized to the cytoplasm, and increased baseline CDK6. RNA-sequencing was performed to gain a global understanding of changes downstream of APC loss and identified a broad mis-regulation of cell cycle-related genes in APC shRNA1 cells. Our studies are the first to show an interaction between APC and taxane response in breast cancer. The implications include designing combination therapy to re-sensitize APC-mutant breast cancers to taxanes using the specific cell cycle alterations.
Article
Full-text available
Paclitaxel administered into the peritoneal cavity is a chemotherapy agent that shows unusually prolonged retention within the peritoneal space. Using this pharmacokinetic fact as a starting point, the use of this drug to benefit patients with peritoneal metastases was investigated. The pharmacokinetics and drug characteristics of paclitaxel were identified from the oncologic literature. The experience to date with ovarian cancer, malignant peritoneal mesothelioma, gastric cancer and pancreas cancer was explored. Paclitaxel given by repeated instillation through an intraperitoneal port has demonstrable responses in ovarian cancer, peritoneal mesothelioma, gastric cancer and pancreas cancer when peritoneal metastases are present. Its role for prevention of peritoneal metastases in patients at high risk seems less well established. Randomized controlled studies have been positive in ovarian cancer but not in other diseases with peritoneal dissemination. A randomized controlled study in gastric cancer with peritoneal metastases produced suggestive but not conclusive results. Conversion surgery after repeated treatments with intraperitoneal paclitaxel has been reported with gastric cancer and pancreas cancer with peritoneal metastases. The pharmacology of intraperitoneal paclitaxel strongly suggest that intraperitoneal administration should be of benefit to prevent or treat peritoneal metastases. Protocols that the oncologist can follow to realize these potential benefits are not as yet available.
Article
Full-text available
Purpose: Appendiceal neoplasms are heterogeneous and are often treated with chemotherapy similarly to colorectal cancer (CRC). Genomic profiling was performed on 703 appendiceal cancer specimens to compare the mutation profiles of appendiceal subtypes to CRC and other cancers, with the ultimate aim to identify potential biomarkers and novel therapeutic targets. Methods: Tumor specimens were submitted to a Clinical Laboratory Improvement Amendments-certified laboratory (Foundation Medicine, Cambridge, MA) for hybrid-capture-based sequencing of 3,769 exons from 315 cancer-related genes and 47 introns of 28 genes commonly rearranged in cancer. Interactions between genotype, histologic subtype, treatment, and overall survival (OS) were analyzed in a clinically annotated subset of 76 cases. Results: There were five major histopathologic subtypes: mucinous adenocarcinomas (46%), adenocarcinomas (30%), goblet cell carcinoids (12%), pseudomyxoma peritonei (7.7%), and signet ring cell carcinomas (5.2%). KRAS (35% to 81%) and GNAS (8% to 72%) were the most frequent alterations in epithelial cancers; APC and TP53 mutations were significantly less frequent in appendiceal cancers relative to CRC. Low-grade and high-grade tumors were enriched for GNAS and TP53 mutations, respectively (both χ2P < .001). GNAS and TP53 were mutually exclusive (Bonferroni corrected P < .001). Tumor grade and TP53 mutation status independently predicted OS. The mutation status of GNAS and TP53 strongly predicted OS (median, 37.1 months for TP53 mutant v 75.8 GNAS-TP53 wild type v 115.5 GNAS mutant; log-rank P = .0031) and performed as well as grade in risk stratifying patients. Conclusion: Epithelial appendiceal cancers and goblet cell carcinoids show differences in KRAS and GNAS mutation frequencies and have mutation profiles distinct from CRC. This study highlights the benefit of performing molecular profiling on rare tumors to identify prognostic and predictive biomarkers and new therapeutic targets.
Article
Full-text available
Appendiceal adenocarcinoma (AA) is an orphan disease with unique clinical attributes but often treated as colorectal cancer (CRC). Understanding key molecular differences between AA and CRC is critical. We performed retrospective analyses of AA patients (N = 266) with tumour and/or blood next-generation sequencing (NGS) (2013–2018) with in-depth clinicopathological annotation. Overall survival (OS) was examined. For comparison, CRC cohorts annotated for sidedness, consensus molecular subtypes (CMS) and mutations (N = 3283) were used. Blood-NGS identified less RAS/GNAS mutations compared to tissue-NGS (4.2% vs. 60.9%, P < 0.0001) and showed poor concordance with tissue for well-/moderately differentiated tumours. RAS (56.2%), GNAS (28.1%) and TP53 (26.9%) were most frequent mutations. Well/moderately differentiated tumours harboured more RAS (69.2%/64.0% vs. 40.5%) and GNAS (48.7%/32.0% vs. 10.1%) while moderate/poorly differentiated tumours had more TP53 (26.0%/27.8% vs. 7.7%) mutations. Appendiceal adenocarcinoma (compared to CRC) harboured significantly fewer APC (9.1% vs. 55.4%) and TP53 (26.9% vs. 67.5%) and more GNAS mutations (28.1% vs. 2.0%) (P < 0.0001). Appendiceal adenocarcinoma mutation profile did not resemble either right-sided CRC or any of the four CMS in CRC. High grade, but no mutation, was independently predictive of survival. Integrated clinico-molecular profiling of AA identified key molecular drivers distinct from CRC. Appendiceal adenocarcinoma has a predominantly grade-driven biology that trumps mutations.
Article
Appendiceal adenocarcinomas (AA) are a rare and heterogeneous mix of tumors for which few preclinical models exist. The rarity of AA has made performing prospective clinical trials difficult, which has partly contributed to AA remaining an orphan disease with no chemotherapeutic agents approved by the FDA for its treatment. AA has a unique biology in which it frequently forms diffuse peritoneal metastases but almost never spreads via a hematogenous route and rarely spreads to lymphatics. Given the localization of AA to the peritoneal space, intraperitoneal delivery of chemotherapy could be an effective treatment strategy. Here, we tested the efficacy of paclitaxel given by intraperitoneal administration using three orthotopic patient-derived xenograft (PDX) models of AA established in immunodeficient NSG mice. Weekly intraperitoneal paclitaxel treatment dramatically reduced AA tumor growth in all three PDX models. Comparing the safety and efficacy of intravenous with intraperitoneal administration, intraperitoneal delivery of paclitaxel was more effective, with reduced systemic side effects in mice. Given the established safety record of intraperitoneal paclitaxel in gastric and ovarian cancers, and lack of effective chemotherapeutics for AA, these data showing the activity of intraperitoneal paclitaxel in orthotopic PDX models of mucinous AA support the evaluation of intraperitoneal paclitaxel in a prospective clinical trial. Significance The activity and safety of intraperitoneal paclitaxel in orthotopic PDX models of mucinous appendiceal adenocarcinoma supports the evaluation of intraperitoneal paclitaxel in a prospective clinical trial of this rare tumor type.
Article
Purpose: Appendiceal adenocarcinomas (ACs) are rare, histologically diverse malignancies treated as colorectal cancers despite having distinct biology and clinical behavior. To guide clinical decision making, we defined molecular subtypes of AC associated with patient survival, metastatic burden, and chemotherapy response. Patients and methods: A comprehensive molecular analysis was performed in patients with AC to define molecular subtypes. Associations between molecular subtype and overall survival, intraoperative peritoneal cancer index, and first-line chemotherapy response were assessed adjusting for histopathologic and clinical variables using multivariable Cox proportional hazards, linear regression, and logistic regression models. Results: We defined distinct molecular lineages of mucinous appendiceal adenocarcinoma (MAAP) from co-occurring mutations in GNAS, RAS, and TP53. Of 164 MAAP tumors, 24 were RAS-mutant (mut) predominant (RAS-mut/GNAS-wild-type [wt]/TP53-wt) with significantly decreased mutations and chromosomal alterations compared with tumors with GNAS mutations (GNAS-mut predominant) or TP53 mutations (TP53-mut predominant). No patient with RAS-mut predominant subtype metastatic MAAP died of cancer, and overall survival in this subgroup was significantly improved compared with patients with GNAS-mut (P = .05) and TP53-mut (P = .004) predominant subtypes. TP53-mut predominant subtypes were highly aneuploid; increased tumor aneuploidy was independently (P = .001) associated with poor prognosis. The findings retained significance in patients with any metastatic AC. RAS-mut predominant metastases exhibited reduced peritoneal tumor bulk (P = .04) and stromal invasion (P < .001) compared with GNAS-mut or TP53-mut predominant tumors, respectively. Patients with RAS-mut predominant MAAP responded more to first-line chemotherapy (50%) compared with patients with GNAS-mut predominant tumors (6%, P = .03). Conclusion: AC molecular patterns identify distinct molecular subtypes: a clinically indolent RAS-mut/GNAS-wt/TP53-wt subtype; a chemotherapy-resistant GNAS-mut predominant subtype; and an aggressive, highly aneuploid TP53-mut predominant subtype. Each subtype exhibits conserved clinical behavior irrespective of histopathology.
Article
The Chicago Consensus Working Group provides multidisciplinary recommendations for the management of appendiceal neoplasms specifically related to the management of peritoneal surface malignancies. These guidelines are developed with input from leading experts, including surgical oncologists, medical oncologists, pathologists, radiologists, palliative care physicians, and pharmacists. These guidelines recognize and address the emerging need for increased awareness in the appropriate management of peritoneal surface disease. They are not intended to replace the quest for higher levels of evidence. The Chicago Consensus Working Group provides multidisciplinary recommendations for the management of appendiceal neoplasms specifically as it relates to the management of peritoneal surface malignancy. They are not intended to replace the quest for higher levels of evidence.
Article
Currently, treatment of small bowel adenocarcinoma (SBA) mirrors that of colorectal cancer (CRC). Recent genomic data have demonstrated SBA to be a genetically unique entity, suggesting that therapies not traditionally utilized in CRC should be explored. In order to further characterize the activity of taxanes in this rare cancer, we completed a single-center retrospective study. Twenty patients were found to have been treated with taxane-based regimens (monotherapy in 3, combination therapy in 17). Median time to progression was 3.8 months (95% confidence interval [CI] 2.9–4.6), and median overall survival was 10.7 months (95% CI: 3.1–18.3). The results of this study demonstrate clinical activity from taxane-based therapy in advanced SBA and support further clinical trial investigation.
Article
Background: Hypermethylation of promoter CpG islands (CIMP) represents a unique pathway for the development of colorectal cancer (CRC), characterized by lack of chromosomal instability and a low rate of adenomatous polyposis coli (APC) mutations, which have both been correlated with taxane resistance. Similarly, small bowel adenocarcinoma (SBA), a rare tumor, also has a low rate of APC mutations. This phase II study evaluated taxane sensitivity in SBA and CIMP-high CRC. Patients and methods: The primary objective was Response Evaluation Criteria in Solid Tumors version 1.1 response rate. Eligibility included Eastern Cooperative Oncology Group performance status 0/1, refractory disease, and SBA or CIMP-high metastatic CRC. nab-paclitaxel was initially administered at a dose of 260 mg/m2 every 3 weeks but was reduced to 220mg/m2 owing to toxicity. Results: A total of 21 patients with CIMP-high CRC and 13 with SBA were enrolled from November 2012 to October 2014. The efficacy-evaluable population (patients who received at least three doses of the treatment) comprised 15 CIMP-high CRC patients and 10 SBA patients. Common grade 3 or 4 toxicities were fatigue (12%), neutropenia (9%), febrile neutropenia (9%), dehydration (6%), and thrombocytopenia (6%). No responses were seen in the CIMP-high CRC cohort and two partial responses were seen in the SBA cohort. Median progression-free survival was significantly greater in the SBA cohort than in the CIMP-high CRC cohort (3.2 months compared with 2.1 months, P = 0.03). Neither APC mutation status nor CHFR methylation status correlated with efficacy in the CIMP-high CRC cohort. In vivo testing of paclitaxel in an SBA patient-derived xenograft validated the activity of taxanes in this disease type. Conclusion: Although preclinical studies suggested taxane sensitivity was associated with chromosomal stability and wild-type APC, we found that nab-paclitaxel was inactive in CIMP-high metastatic CRC. nab-paclitaxel may represent a novel therapeutic option for SBA.