You are on page 1of 67

Pulmonary Adenocarcinoma:

Approaches to Treatment, 1e Leora


Horn Md Msc
Visit to download the full and correct content document:
https://ebookmass.com/product/pulmonary-adenocarcinoma-approaches-to-treatment
-1e-leora-horn-md-msc/
Pulmonary
Adenocarcinoma:
Approaches to
Treatment
LEORA HORN, MD, MSC
Associate Professor of Medicine
Medicine - Hematology Oncology
Vanderbilt Univeristy
Nashville, TN, United States

]
3251 Riverport Lane
St. Louis, Missouri 63043

Pulmonary Adenocarcinoma: Approaches to Treatment ISBN: 978-0-323-66209-3

Copyright Ó 2019 Elsevier, Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about
the Publisher’s permissions policies and our arrangements with organizations such as the Copyright
Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/
permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices

Practitioners and researchers must always rely on their own experience and knowledge in evaluating
and using any information, methods, compounds or experiments described herein. Because of rapid
advances in the medical sciences, in particular, independent verification of diagnoses and drug
dosages should be made. To the fullest extent of the law, no responsibility is assumed by Elsevier,
authors, editors or contributors for any injury and/or damage to persons or property as a matter of
products liability, negligence or otherwise, or from any use or operation of any methods, products,
instructions, or ideas contained in the material herein.

Publisher: Dolores Meloni


Acquisition Editor: Robin R Carter
Editorial Project Manager: Jennifer Horigan
Production Project Manager: Poulouse Joseph
Designer: Alan Studholme
List of Contributors

Leora Horn, MD, MSc Michael J. Jelinek, MD


Associate Professor of Medicine Hematology/Oncology Fellow
Medicine e Hematology Oncology Department of Medicine
Vanderbilt Univeristy University of Chicago
Nashville, TN, United States Chicago, IL, United States

Erin A. Gillaspie, MD, MPH Stephen V. Liu, MD


Assistant Professor of Thoracic Surgery Associate Professor of Medicine
Thoracic Surgery Division of Oncology
Vanderbilt University Medical Center Georgetown University
Nashville, TN, United States Lombardi Comprehensive Cancer Center
Washington, DC, United States
Ming-Sound Tsao, MD, FRCPC
Bhavisha A. Patel, MD
Professor
Hematology/Oncology Fellow
Laboratory Medicine and Pathobiology
Department of Hematology/Oncology
University of Toronto
Medstar Washington Hospital Center
Consultant Pathologist and Senior Scientist
Washington, MD, United States
Princess Margaret Cancer Centre
Toronto, ON, Canada
Lecia V. Sequist, MD, MPH
The Landry Family Associate Professor of Medicine
Heather A. Wakelee, MD Harvard Medical School
Professor of Medicine, Oncology Boston, MA, United States
Stanford University/Stanford Cancer Institute
Director
Stanford, CA, United States
Center for Innovation in Early Cancer Detection
Massachusetts General Hospital Cancer Center
Jeffrey R. Zweig, MD Boston, MA, United States
Fellow in Hematology/Oncology
Department of Medicine Rina Hui, MBBS, PhD
Divisions of Hematology and Oncology Associate Professor
Stanford Cancer Institute Department of Medical Oncology
Stanford University Westmead Hospital and the University of Sydney
Stanford, CA, United States Sydney, NSW, Australia

Hak Choy, MD J. Travis Mendel, MD


Chair Resident
Radiation Oncology Radiation Oncology
UT Southwestern UT Southwestern
Dallas, TX, United States Dallas, TX, United States

v
vi LIST OF CONTRIBUTORS

Shirish Gadgeel, MD Dan Zhao, MD, PhD


Professor Fellow
Department of Internal Medicine, Division Hematology/Oncology
of Hematology & Oncology City of Hope Comprehensive Cancer Center
University of Michigan Duarte, CA, United States
Ann Arbor, MI, United States
Prodipto Pal, MD, PhD
Jyoti D. Patel, MD, FASCO Assistant Professor
Professor of Medicine Thoracic Pathology Service
University of Chicago Department of Laboratory Medicine and Pathobiology
Chicago, IL, United States University Health Network - University of Toronto
Toronto, ON, Canada
Michael Millward, MBBS, MA
Professor Michael Cabanero, MD
Cancer Council Professor of Clinical Cancer Research Assistant Professor
Consultant Medical Oncologist Thoracic Pathology Service
Sir Charles Gairdner Hospital and the University of Department of Laboratory Medicine and Pathobiology
Western Australia University Health Network - University of Toronto
Perth, WA, Australia Toronto, ON, Canada

Emily Dickinson Nicolas Marcoux, MD, FRCPC


Student Research Fellow
Cognitive Sciences Massachusetts General Hospital Cancer Center
Rice University Boston, MA, United States
Houston, TX, United States Medical Oncologist, Hematologist
CHU de Québec
Karen L. Reckamp, MD, MS
Quebec City, Canada
Professor
Department of Medical Oncology
City of Hope Comprehensive Cancer Center
Duarte, CA, United States
Preface

Lung cancer is the leading cause of cancer-related mor- The majority of patients, however, will present with
tality worldwide. The past three decades have seen the advanced stage disease that may be treatable but not
therapeutic approach to lung cancer evolve from a tactic curable with current systemic therapies. We have seen
of one size fits all to classifying the tumor into small cell rapid advances made in the treatment landscape for
lung cancer or nonesmall cell lung cancer (NSCLC) patients with tumors that harbor the EGFR or BRAF
based on immunohistochemical stains. Approximately mutation and the ALK or ROS1 fusion. For this cohort of
85% of patients with lung cancer will have NSCLC, patients, which account for less than 20% of North
which can be further subclassified into adenocarcinoma, American patients with advanced stage disease, tyrosine
squamous cell carcinoma, or large cell neuroendocrine kinase inhibitors (TKIs) are approved as first-line therapy
histology. based on studies that have demonstrated an improved
All histologic types of lung cancer can develop in response rate, progression-free survival (PFS), and over-
current and former smokers. However, with the decline all survival (OS) compared with chemotherapy or his-
in cigarette consumption, adenocarcinoma has become torical controls. Moreover, for patients with tumors that
the most frequent histologic subtype of lung cancer are HER2, MET, RET, and NTRK positive, each account-
diagnosed in the United States. It also tends to be the ing for less than 2% of NSCLC adenocarcinoma, there
most common form of lung cancer diagnosed in lifetime are exciting agents in clinical development that will
never smokers or former light smokers (<15 pack-year further expand on oral treatment options for patients
history), women, and younger adults (<60 years). with NSCLC adenocarcinoma histology. For the majority
A diagnosis of adenocarcinoma alone has become of patients with no oncogenic driver, or for a driver for
insufficient to make therapeutic recommendations. The which there is no targeted therapy at this time, such as
identification of actionable driver mutations and the 25% of patients with KRAS mutation positive
knowledge of to programmed death ligand 1 (PD-L1) NSCLC, chemotherapy for many years was the standard
status has changed the landscape of treatment options of care with important patient considerations required in
and trajectory of outcomes for a cohort of patients, and selecting the optimal first- and second-line treatment
testing patients at diagnosis has become the standard of option. With the introduction of immune checkpoint
care. inhibitors into the treatment armamentarium, we have
In this book, we describe the optimal treatment seen rapid improvements in outcomes for patients with
approach to patients with adenocarcinoma of the lung advanced stage disease.
from diagnosis to end-of-life care. We describe the Adenocarcinoma of the lung provides a comprehen-
appropriate testing required to make a pathologic diag- sive overview of the rapidly evolving treatment paradigm
nosis of NSCLC adenocarcinoma histology, including for patients with NSCLC, providing a valuable resource
molecular and PD-L1 testing. We also review the optimal to all interested in this disease.
surgical approaches and adjuvant therapy recommen-
dations for patients diagnosed with early-stage disease Leora Horn
and radiation approaches for patients with locally July 2018
advanced disease.

vii
CHAPTER 1

The Surgical Management of Pulmonary


Adenocarcinoma
ERIN A. GILLASPIE, MD, MPH

INTRODUCTION PET scan abnormalities with tissue diagnosis remains


The management of lung cancer has evolved dramati- important as the imaging also carries a significant rate
cally over the last two decades. Importantly, research of false-positive upstaging in particular regions where
has established that a multidisciplinary approach is histoplasmosis is prevalent.1,2
essential to help achieve optimal long-term outcomes. Imaging and staging should ideally be completed as
Surgery plays an important role in early, locally quickly as possible and should not be any older than
advanced, and select advanced stages of lung cancer. 60 days prior to the initiation of therapy or there is a
While surgical approach continues to transition to risk of progression in the interim.
more and more minimally invasive options, the princi-
ples remain the same. Pathologic Staging: Tissue Diagnosis for
This chapter will focus on establishing patient candi- Suspected Lung Cancer
dacy and surgical considerations for the treatment of Tissue diagnosis may be accomplished through a range
pulmonary adenocarcinoma. of procedures including conventional bronchoscopy,
CT-guided transthoracic needle biopsy, navigational
bronchoscopy, and resectional biopsy (surgery). Resec-
STAGING tional biopsy has largely been supplanted by naviga-
The accurate staging of lung cancer is crucial in helping tional bronchoscopy, which is less invasive than
to guide treatment recommendations. This often endobronchial methods.3
includes a combination of imaging modalities and bi-
opsies to determine size, location, nodal involvement, Endobronchial and Transbronchial Biopsy
and histology. Even with a thorough preoperative Endobronchial and transbronchial approaches to bi-
workup there will be a subset of patients who will opsy are minimally invasive with low morbidity and
have unanticipated findings at the time of surgery. low cost.
Biopsies are obtained through a flexible, fiber-optic
Initial Staging: Imaging bronchoscope. Generally, four to five pieces of tissue
Computed tomography (CT) scanning is a standard are collected, each measuring approximately
part of the workup for lung cancer and in determining 1e2 mm in diameter. Samples may be submitted as
the candidacy for resection. While excellent in delin- a frozen section or in formalin fixative for permanent
eating the extent of primary disease and some sites of section analysis. Although the procedure is low risk,
metastatic disease, it is the least sensitive and specific a careful physical history should be obtained ahead
modality for the identification of lymph node of time to ensure no anesthetic or bleeding risks exist
involvement.1 for a patient.
The utility of positron emission tomography (PET) Endobronchial lesions are visible within the bron-
scan has evolved over time and now holds an important chial tree and may be sampled directly using a biopsy
role in extrathoracic staging to rule out distant forceps though a flexible or rigid bronchoscope.
metastasis which would in many cases render a patient Transbronchial biopsy is also performed with the
unresectable. PET is also helpful in assessing the medi- assistance of a flexible bronchoscopy. At minimum, a
astinum for lymph node involvement. Confirmation of chest X-ray is required, and preferably, a CT scan is

Pulmonary Adenocarcinoma: Approaches to Treatment. https://doi.org/10.1016/B978-0-323-55433-6.00001-8


Copyright © 2019 Elsevier Inc. All rights reserved. 1
2 Pulmonary Adenocarcinoma: Approaches to Treatment

performed prior to the procedure to help guide tissue diagnostic yielded has been demonstrated to be
sampling. Fluoroscopy is commonly used intraproce- equivalent.4,13,14
durally to help localize a target lesion to improve accu- An open lung biopsy allows surgeons to palpate and
racy of the diagnostic yield. The most common visualize the whole lung and directly select the lesions
complication associated with the biopsy of peripheral to biopsy. Palpating lesions is more challenging in a
nodules is pneumothorax.4 VATS approach. For small lesions or ground-glass opac-
Electromagnetic navigational bronchoscopy (ENB) ities where identification can be difficult, localization
is a newer technology that allows a practitioner to may be performed preoperatively to assist using ENB
use CT scan to plot a course through the bronchial to inject blue dye or place a fiducial prior to thoraco-
tree to a lung nodule or desired biopsy location.5 scopy. An alternative and equally effective method as
This technology is particularly effective in more described in a paper by Grogan et al. is preoperative
difficult-to-reach nodules and smaller nodules. Zhang CT-guided radionucleotide labeling of a lesion with
et al. performed a metaanalysis to evaluate the overall technetium 99. Intraoperatively, a collimated probe
diagnostic yield and accuracy of ENB and a pooled connected to a g detector can be used to isolate the
sensitivity of 82% and specificity of 100%.6 In a meta- lesion. The lesion may be wedged out, and then back-
analysis, Gex et al. identified the variables favorably ground counts can then be assessed to confirm spec-
influencing the performance of ENB include greater imen removal. Localization has been described to be
size of the nodule, nodule visualization using a successful in 95% of cases.15
radial-probe EBUS, presence of a bronchus sign, lower An open lung biopsy specimen may be reviewed
registration error, and catheter suction technique. intraoperatively, and if malignancy is confirmed, a
Overall, the technique has been demonstrated to be definitive resection can be performed in the same
safe and effective.7 setting if appropriate. Surgical considerations will be
Fine needle aspiration (FNA) and needle biopsy are discussed in more detail later in the chapter.
alternative methods for sampling nodules.8 CT scan is
the imaging modality of choice to visualize the needle Pathologic Mediastinal Assessment
advancing within the nodule or area of abnormality.9 Imaging can be informative and is an important first
The accuracy for CT-guided biopsies range from 64% step in the assessment of mediastinal nodes. CT has a
to 97%. A trend toward lower diagnostic accuracy was sensitivity of 55% and specificity of 81%, while PET-
noted for smaller lesions less than 1.5 cm in diameter CT has a sensitivity of 62% and specificity of 90%.16
and in a subpleural location.8 The most common Mediastinoscopy has long been considered the gold
complications are pneumothorax and hemorrhage. standard diagnostic technique for the assessment of
Risk factors for complications include smaller size of mediastinal lymph nodes. Mediastinoscopy can eval-
lesions, length of intrapulmonary needle path, and uate level 2, 4, and 7 nodes and can send large tissue
trans-fissure course.10,11 samples for analysis. This has been replaced in many
In 2014, Capalbo et al. compared FNA and core nee- centers by endobronchial ultrasound (EBUS) as the first
dle biopsy in 121 of their patients. FNA resulted in line. Despite this, mediastinoscopy does maintain an
fewer complicationsdpneumothorax in 18% versus important role in diagnosis of suspicious lymphade-
31% and parenchymal hemorrhage in 9% versus nopathy or mediastinal restaging and carries a sensi-
34%dand diagnostic accuracy of FNA was 94.8% tivity of 89% and specificity of 100%.1
when performed in the presence of a pathologist who EBUS with transbronchial needle aspiration has
could confirm adequacy of tissue sampling.12 increasingly been used as a favored method for
diagnosis of suspicious mediastinal nodes. EBUS is
Surgical Lung Biopsy unique in that it offers the ability to sample both N1
Open lung biopsy has taken on new meaning in the and N2 nodes. Caution should be exercised when
minimally invasive surgical era. Standard video- considering resectability for left upper lobe tumors as
assisted thoracoscopic surgery (VATS) and uniportal they have the most unpredictable pattern of lymph
VATS have replaced open thoracotomy as the approach node metastasis and frequently involve level 5 and 6
of choice. nodes which are inaccessible to mediastinoscopy and
Early in the experience, outcomes were compared EBUS. Review of available publications reveals a me-
between diagnostic efficacies of thoracotomy and dian sensitivity of 89% and has a very low risk of
VATS approach. No matter the surgical approach, complications.1,17,18
CHAPTER 1 The Surgical Management of Pulmonary Adenocarcinoma 3

Rapid onsite cytologic evaluation confirms adequacy essential to ensure adequate pulmonary reserve prior
of samplings at the time of procedure to enhance over- to proceeding with resection.
all yield.19 Spirometry and measurement of diffusion capacity
The number of lymph node stations to assess is for carbon monoxide (DLCO) are recommended for
debated among experts with some favoring the routine all patients undergoing pulmonary resection and are
sampling of all lymph node stations while others favor- calculated as a percentage of normal.28
ing only the sampling of those nodes that are suspicious The forced expiratory volume in 1 s (FEV1),
on imaging.18,20 measured during spirometry, determines the mechani-
Even with negative imaging, negative surgical cal function of the lung, i.e., the airflow limitations of
biopsy, and a small (T1) cancer, there will still be a medium to large airways. DLCO value provides a gross
subset of patients found to have occult N2 disease estimate of the function of the alveolar-capillary
either on frozen section at the time of lung resection membrane.
or discovered later on final pathology. The rate of A preoperative predicted value of 60% or less for
occult N2 disease with T1 primary tumors and negative FEV1 has been shown to be an important cut-off value
imaging is in the range of 6.1%e6.5% and 8.7% for T2 for predicting respiratory complications.29 DLCO is a
tumors.21,22 Although prediction models to try to second, independent functional parameter that can
determine the risk of N2 disease have been developed separately predict surgical risk and is currently ordered
and tested, they are not widely accepted and as a standard part of workup along with pulmonary
adopted.23,24 function testing. If this is also less than 60%e70%
on preoperative predictive value, additional testing
is warranted to help determine the safety of
ASSESSING SURGICAL CANDIDACY resection.30e32
After establishing the stage of cancer, a meticulous A postoperative predicted value of DLCO and FEV1
preoperative evaluation should be performed prior to may be calculated directly by subtracting the number
recommending resection. Workup should include his- of segments that would be resected at the time of
tory and physical examination, pulmonary function surgery, assuming that each contributes equally. A
tests, a nutritional assessment, and cardiac clearance. more accurate method would be to obtain a quantita-
tive lung scan to directly determine the contribution
Smoking Cessation from each portion of the lung.27,32
Smokers should be counseled to stop immediately. In a DLCO postoperative predicted value of less than
study by Mason et al. in 2009 from the General Thoracic 60% in some studies and 40% in others denotes a sig-
Surgery Database, the risk of in-hospital death and pul- nificant risk for perioperative morbidity and mortality.
monary complications was higher in patients who were For FEV1 values of less than 40% and DLCO of less
actively smoking at the time of surgery and could be than 40%, exercise stress testing should be
mitigated by preoperative smoking cessation.25 performed.32e34
Options for secondary testing include a shuttle walk
Assessment of Cardiovascular Risk test, stair climbing, exercise stress test or a perfusion
Major cardiac-related adverse events occur in approxi- scan. The former are more subjective, while later are
mately 2%e3% of patients after pulmonary resection. more quantitative and have established thresholds asso-
The thoracic revised cardiac risk index is a validated ciated with surgical risk.
tool that can be used to help guide which patients A quantitative radionucleotide scan uses inhaled
should be sent for additional, preoperative consulta- radioactive xenon and the intravenous administration
tion. Patients who score more than 1.5 in the cardiac in- of technetium-labeled macroaggregates to determine
dex or those describing limited exercise capacity or a the fraction of total perfusion for the upper, middle,
recent diagnosis of active heart disease all warrant and lower zones of the lung.35
evaluation.26,27 Cardiopulmonary exercise testing is a physiologic
testing technique that provides an evaluation of func-
Assessment of Pulmonary Function tional capacity of both the heart and lungs.
Patients undergoing pulmonary resection commonly The risk for perioperative complications has been
have a number of coexisting conditions and exposures reported to be higher with the lower measured value
which can perturb pulmonary function. It is therefore of VO2 max. Values over 20 mL/kg min can safely
4 Pulmonary Adenocarcinoma: Approaches to Treatment

undergo a planned resection. A value between 10 and T3N1 Tumors


15 mL/kg min indicates an increased risk of periopera- T3N1 is a relatively infrequent stage of lung cancer but
tive death.27,36 Brunelli et al. observed a mortality rate surgery has a well-defined role as the primary treatment
of 13% with a VO2 max of <12%, while no mortality for these tumors. T3 lung cancers range in size from 5 to
was observed with >20.36 7 cm or invade structures that can be routinely taken en
Licker et al. found in their study that VO2 max bloc with the lungdthe chest wall, pericardium, or a
of <10 mL/kg min is at very high risk and considered single phrenic nerve. Often, hilar nodal involvement
a contraindication to anatomic resectiondtotal is discovered at the time of resection. The surgical goal
morbidity 5%, cardiac morbidity 39%.29 is an en bloc resection with microscopically negative
margins.39,40
The most common presentation is with chest wall
SURGICAL MANAGEMENT OF LUNG involvement. Chest wall resection can be performed
CANCER en bloc with a pulmonary resection. At least 2e3 cm
Stage, histology, and patient candidacy determine the margins or one rib above and below the involved
treatment modalities that will confer the best overall area should be included in the resection to achieve a
survival. Surgery plays an important role in the manage- complete microscopic resection. As thoracoscopic
ment of lung cancer, and this approach has evolved equipment has evolved and thoracic surgeon experience
significantly over the last two decades. has increased, more complex cases are able to be
completed minimally invasively. Small chest wall de-
Early-Stage Lung Cancer fects and those hidden by the scapula do not require
Surgery is considered to be a standard part of the treat- reconstruction. A defect greater than 5 cm should be
ment for stage I and II nonesmall cell lung cancers reconstructed along with resection of the fifth to sixth
(NSCLCs) (Table 1.1). The eighth edition staging ribs near the tip of the scapula to prevent entrapment
system comprehends tumors that are node negative and to maintain chest wall integrity.
and up to 7 cm in size and tumors that involve the chest There is currently no clear evidence for neoadjuvant
wall, pericardium, or satellite nodules in the same lobe. chemotherapy in these patients; however, adjuvant
Surgery is also performed for patients who have smaller chemotherapy is recommended for tumors that are
tumors up to 5 cm or involving the main bronchus, determined on final pathology to be greater than 5 cm
ipsilateral, hilar nodes.37,38 or have positive ipsilateral hilar lymph nodes. Postoper-
ative radiation is usually reserved for patients in whom
Locally Advanced Lung Cancer there is a concern for residual disease.39,40
The treatment of locally advanced lung cancer is com-
plex and requires a multidisciplinary approach. The N2 Disease
presentations in which surgery can play a role in therapy Approximately, 15% of patients with pulmonary
are described below.39 adenocarcinoma will present with stage IIIA (N2) dis-
ease. The optimal management of N2 disease is
perhaps the most controversial in thoracic oncology.
Ipsilateral mediastinal lymph node involvement
TABLE 1.1
ranges from occult disease to single station, micro-
Early-Stage Lung Cancer scopic disease to multistation bulky disease, and every-
N0 N1 N2 N3 thing in between.
T1 IA IIB IIIA IIIB Even with negative imaging and negative pathologic
mediastinal staging, there is a subset of patients who
T2a IB IIB IIIA IIIB
are discovered to have N2 disease at the time of lung
T2b IIA IIB IIIA IIIB resection. The rate ranges from 6.1% to 8.7%.21,22
T3 IIB IIIA IIIB IIIC When confronted with unseen N2 disease at the time
T4 IIIA IIIA IIIB IIIC of surgery, many surgeons will complete resection of
the tumor and refer the patient for consideration
Adapted from Detterbeck et al. Eighth edition lung cancer staging. of adjuvant therapy as the patient has already been sub-
jected to the risk of anesthesia and thoracic surgery.
CHAPTER 1 The Surgical Management of Pulmonary Adenocarcinoma 5

For preoperatively identified N2 disease, there is no Surgery should be performed in high, volume
consensus among surgeons as to what defines resectable thoracic centers. Meticulous preoperative planning
N2 disease and who should be considered for surgery as and in some cases multidisciplinary surgical approach
part of a multimodal treatment regimen. However, the is necessary to ensure complete resection.42
INT0139 trial firmly established that surgery alone was Due to the heterogeneity of this population, it is
insufficient for the management of N2 disease. difficult to accumulate sufficient evidence to clearly
As a part of a multimodal treatment regimen, studies define the role of surgery in this group of patients.
suggest that surgery can enhance disease-free and over-
all survival in patients who do not present with bulky Superior Sulcus Tumors
mediastinal disease, those who are able to complete Superior sulcus tumors, also known as Pancoast tu-
neoadjuvant therapy and pathologic partial or complete mors, arise from the apex of the right upper or left upper
response at the time of resection.21,22 lobe. These tumors may invade any of the structures in
Additional research is required to help establish a the thoracic inlet (Fig. 1.1).
more structured treatment plan for patients presenting As with other locally advanced tumors, multidisci-
with N2 disease. plinary approach is the mainstay of management of su-
perior sulcus tumors with chemoradiotherapy
T4 Tumors administration followed by restaging to determine
T4 tumors are locally aggressive with invasion of critical resectability.
mediastinal structures such as the PA, heart, aorta, supe- The factors associated with poor prognosis include
rior vena cava (SVC), trachea, esophagus, spine, or the incomplete resection, lack or response to neoadjuvant
diaphragm. In many institutions, these advanced-stage treatment, invasion into the brachial plexus, >50% of
tumors are considered unresectable and relegated to the vertebral body and great vessels as well as lymph
chemotherapy and radiation. node involvement.43
Several nonrandomized series have shown that high-
ly selected patients may have 5-year survival rates as Oligometastatic Disease
high as 48%. Most reported experience involves tumors The concept of oligometastatic disease was first pro-
invading the SVC, left atrium, carina, intrapericardial posed in 1995 and then further defined in 2006 by
pulmonary vessels, and vertebral bodies. Prognosis is Niibe et al., who classified oligometastatic disease by
associated with achieving en bloc survival and limited prognosis. They described oligorecurrence or oligome-
or no nodal involvement (N0eN1).41e43 tastatic disease in the adrenal or brain as actually having

A B
FIG. 1.1 Right-sided superior sulcus tumor seen in coronal (A) and axial (B) views. The tumor is invading the
posterior chest wall.
6 Pulmonary Adenocarcinoma: Approaches to Treatment

a relatively favorable prognosis. For these lesions, the being performed minimally invasively with this
standard of care includes surgical resection of both the approach being available in 80% of hospitals across
primary and metastasis or recurrence.44,45 Generally the nation.49,50
for synchronous disease, the metastatic site is treated A VATS lobectomy is conducted in similar fashion to
first. If this is not able to be successfully addressed, there an open lobectomy but utilizes 2e3 small ports in the
is no benefit to resecting the primary. chest, each measuring approximately 1 cm, and a
slightly larger utility incision measuring 3 cm that is
used to remove the specimens (Fig. 1.2). VATS differs
SURGICAL APPROACH TO LUNG CANCER from open surgery in both the length of incisions and
The conduct of the operation begins with the adminis- that there is minimal muscular division and no rib
tration of anesthesia and placement of an endotracheal spreading (Fig. 1.3).
tube that allows for pulmonary isolation. This may be A camera allows visualization of the whole hemi-
accomplished with a double-lumen endotracheal tube thorax and special instruments have been designed to
or a bronchial blocker. allow for uses a camera to provide visualization of the
The approach to surgery and extent of pulmonary whole chest and special instruments to facilitate.
resection is determined by the size and location of the Comparison of the outcomes of VATS versus open
tumor and experience of the surgeon. lobectomies have shown VATS to be a safe surgery
with oncologically equivalent outcomes.46,51,52 Both
Open Lobectomy VATS and open surgery have similar patterns of
The majority of pulmonary resections continue to be
performed in open fashion. An open lobectomy is per-
formed through a thoracotomy: axillary, anterolateral,
or posterolateral.
A posterolateral thoracotomy is the most standard
approach and is performed using a semi-muscle-sparing
approach by dividing the latissimus and sparing the ser-
ratus anterior muscle. If the latissimus can be retracted,
rather than divided, then this is preferred.
The chest is entered and a rib can be shingled to
assist with exposure. The lung is rendered atelectatic,
and the chest and lung are inspected for unexpected A
findings and to confirm resectability.
Surgery begins with the removal of N1 and N2
lymph nodes which are sent to pathology. A lobectomy
is performed by isolating the arteries, vein, and bron-
chus associated with the lobe and dividing each, along
with the fissure that connects the upper, middle (right),
and lower lobes. Once the lobectomy is completed, the
remaining lung is reinflated to fill the pleural space, and
chest tubes are positioned to help drain fluid and air
while the patient recovers.
Lung resections carry a significant risk of periopera-
tive morbidity with 37% of patients experiencing
some form of postoperative complicationsdatrial B
arrhythmia and prolonged air leak being the most com-
mon. Other complications can include infection, FIG. 1.2 Isolation of a branch of the left upper lobe
bleeding, hypoxia, and death.46 pulmonary artery (A) and left upper lobe pulmonary vein (B)
during a left upper lobectomy. (A) The anatomy of the left
Video-Assisted Thoracoscopic Surgery upper lobe pulmonary artery is highly variable. The branches
are dissected out and divided with serial applications of a
Lobectomy
vascular load stapling device. (B) The superior pulmonary
The first reports of VATS lobectomy were in 1992 and vein drains the upper lobe of the lung. The vein is isolated, but
1993.47,48 Adoption across the United States has not divided until a separate inferior pulmonary vein is
been slow, but now approximately 44% of cases are confirmed.
CHAPTER 1 The Surgical Management of Pulmonary Adenocarcinoma 7

postoperative complications, although VATS occurs at a Much like VATS, robotic lobectomy is performed
lesser rate.53e55 Additional benefits include earlier re- through small (8 mm) incisions with a slightly different
covery, less pain, less impact on pulmonary function, configuration (Fig. 1.4).
better quality of life, and increased delivery of adjuvant This newer technology allows several advantages
therapy.56e60 over traditional thoracoscopic surgery in that it provides
Despite the evidence to support benefits to patients, a three-dimensional high-definition field and the in-
VATS lobectomy has had variable uptake across the struments have 9 degrees of freedom and include the
United States with some institutions performing only ability to stable and seal vessels.
open procedures while others are performing in excess Studies have demonstrated equal safety and opera-
of 90% of cases thoracoscopically. The ideal proportion tive times with comparable morbidity, mortality, and
of VATS lobectomy is not yet well defined. There benefits to VATS including survival. Cost studies have
equally is no great explanation for the significant demonstrated that while upfront cost is more, ulti-
variability in practice patterns. Initial safety concerns mately minimally invasive surgery including robotic
have been put to rest and excellent training pathways surgery ultimately provide cost savings both intra-oper-
have been developed to help with the adoption of atively and post-operatively.60,63e66
new technology.46 As with any new technology, there is a learning curve
Alternative methods to perform minimally invasive but the da Vinci robots (Intuitive Surgical, Sunnyvale,
surgery have emerged since the advent of VATS lobec-
tomy, and this may help to increase the overall volume
of minimally invasive surgery.

Robotic Lobectomy
Robotic lobectomies were first performed by Morgon
et al. and Ashton et al. in 2003, and the use of this tech-
nology has continued to increase with time.61,62

B
FIG. 1.4 Robotic lobectomy port sites (A) and docking of
the robotic arms (B). (A) Four incisions were created to
FIG. 1.3 Incision for video-assisted thoracoscopic surgery perform the robotic lobectomy, each measuring 8 mm. At the
lobectomy. The incisions are generally placed in the fifth end of the case, the anteriormost incision is lengthened
intercostal space, anterior axillary line, and the eighth slightly to allow the removal of the lobe. A separate incision is
intercostal space anterior and posterior axillary line. There used for the chest tube. (B) Once the ports are placed, the
are, however, variations depending on the lobe that is being robotic arms are docked, and instruments are inserted into
resected and surgeon preference. (Photo credit Dominic M the chest. The surgeon can then sit at the console and
Doyle, Ms for Vanderbilt.) perform the surgery using the robotic instruments.
8 Pulmonary Adenocarcinoma: Approaches to Treatment

CA, USA) company has developed comprehensive resections (including segmentectomy or wedge resec-
training programs and certification processes including tion) were thought to be associated with higher risk of
on-site proctoring for cases. recurrence and historically were reserved for patients
with limited cardiopulmonary reserve.
Uniportal and Subxiphoid Lobectomy The advent of CT screening for lung cancer
Uniportal lobectomy is single-incision VATS lobec- and improvements in imaging technology have led
tomy, which became another natural extension of to the identification of a large number of patients
VATS lobectomy. Developed by Dr. Rocco and Dr. with small (<2 cm) peripheral nodules. This promp-
Gondolas-Rivas, this procedure required improvement ted a renewed interest in the possibility of more
in articulating staplers and the development of other limited resection.
reticulating instruments to improve angles of dissec- The advantages of sublobar resection include preser-
tion. Uniportal VATS is cited to have better visualization vation of pulmonary function, which can preserve a pa-
and operative conditions similar to an open tient’s ability to undergo subsequent resections and
approach.67 diminished morbidity and mortality.
Subxiphoid is another iteration of uniportal lobec- Current evidence supporting sublobar resection is
tomy with an incision just below the xiphoid rather mostly single institution; retrospective studies and
than in the chest wall. This is to help eliminate postop- many are in patients who have insufficient cardiopul-
erative pain and chest wall numbness that are associ- monary reserve to tolerate a lobectomy. Studies are
ated with instrumentation of the intercostal spaces. A summarized in Table 1.2.
4-cm incision is created in the subxiphoid space, and The factors identified as potentiating higher recur-
the pleural cavity is accessed. The lobectomy is per- rence were larger tumors (stage IB), inadequate margins
formed in an identical fashion to a VATS lobectomy. (less than 1 cm) highlighting the importance of tumor
The procedure is more challenging due to the longer size influencing local recurrence.78
tunnel and more limited view but can be accom- A large American study by the Cancer and Leukemia
plished safely and with significant reduction in post- Group B (CALGB) is currently underway to try to help
operative pain.68 definitively answer the question as to whether a sublo-
bar resection for tumors <2 cm without any lymph
node involvement is oncologically sufficient.
IMPORTANT SURGICAL CONSIDERATIONS
Extent of Resection Extent of Lymph Node Resection
Lobectomy has long been considered the standard of The accuracy of lymph node staging is important to
care and the most optimal oncologic procedure in the help determine stage and thereby guide adjuvant treat-
management of early-stage NSCLC. More limited ment decisions.

TABLE 1.2
Outcomes of Single-Center Studies Lobectomy Versus Sublobar Resection
SURVIVAL (5 YEARS)
Study Size of Study Stage Lobectomy Sublobar
69
Campione et al. 121 patients IA 65 62
Ucar et al.70 34 IA and B 64 70
71
El-Sherif 784 IA HR 1.39 (favoring sublobar resection)
Kilik72 184 IA and B 447 46
73
Koike 223 IA 90 89
Iwasaki74 86 I, II, IIIA 73 70
Cao75 2745 IA and B HR 0.91 (favoring lobectomy)
76
Okada 567 IA and B 89.6 89.1
Altorki77 347 IA 86 85
CHAPTER 1 The Surgical Management of Pulmonary Adenocarcinoma 9

Does removing more nodes in a mediastinal lymph 4. Bensard DD, McIntyre RC, Waring BJ, Simon JS. Compar-
node dissection detect more disease than systematic ison of video thoracoscopic lung biopsy to open lung bi-
sampling of each lymph node station? The answer opsy in the diagnosis of interstitial lung disease. Chest.
may depend on the T stage of the tumor. 1993;103:7651.
5. Wells AU, Hirani N. Interstitial lung disease guideline.
The ACOSOC Z0030 trial demonstrated that in T1
Thorax. 2008;63(1):v1ev58.
and T2 tumors, a complete lymph node dissection 6. Zhang W, Chen S, Dong X, Lei P. Meta-analysis of the diag-
identified occult N2 disease missed on sampling in nostic yield and safety of electromagnetic navigation bron-
4% of cases. It should be noted that this study had choscopy for lung nodules. J Thorac Dis. 2015;7(5):
low utilization of preoperative mediastinal pathologic 799e809.
staging and even PET-CT was not used until the later 7. Gex G, Pralong JA, Combescure C, Seijo L, Rochat T,
part of the study, potentially reducing the accuracy of Soccal PM. Diagnostic yield and safety of electromagnetic
preoperative staging.46 In addition, despite identifying navigation bronchoscopy for lung nodules: a systematic
the occult N2 disease, this did not confer a survival review and meta-analysis. Respiration. 2014;87(2):
benefit to patients. 165e176.
8. Birchard KR. Transthoracic needle biopsy. Semin Intervent
Sentinel lymph node biopsy (SNLB) has been
Radiol. 2011;28(1):L87e97.
occasionally gained traction in thoracic surgery to 9. Yao X, Gomes MM, Tsao MS, Allen CJ, Geddie W,
help guide lymphadenectomy. The hypothesis was Sehkon H. Fine-needle aspiration biopsy versus core-nee-
based on the same rationale that led to the use of dle biopsy in diagnosing lung cancer: a systematic
SNLB for both melanoma and breast cancer. Unfortu- review. Curr Oncol. 2012;19(1):e16ee27.
nately, studies have not justified the use of SLNB for 10. Rizzo S, Preda L, Raimondi S, et al. Risk factors for compli-
NSCLC.79 cations of CT-guided lung biopsies. Radiol Med. 2011;
Hopefully this question will soon be answered by 116(4):548e563.
the Japanese Clinical Oncology Group Trial (JCOG 11. Saji J, Nakamura H, Tsuchida T, et al. The incidence and
1413) which is a randomized phase III clinical trial risk of pneumothorax and chest tube placement after
percutaneous CT-guided lung biopsy: the angle of the nee-
looking at lobe-specific versus systemic nodal dissection
dle trajectory is a novel predictor. Chest. 2002;121(5):
for clinical stage I and II NSCLCs. They plan to enroll 1521e1526.
1700 patients from 44 institutions with primary 12. Capalbo E, Peli M, Lovisatti M, et al. Trans-thoracic
endpoint of overall survival.80 biopsy of lung lesions: FNAB or CNB? Our experience
and review of the literature. Radiol Med. 2014;199(8):
572e594.
CONCLUSION 13. Allen MS, Deschamps C, Jones DM, Trastek VF,
Surgery continues to play an important role in the man- Pairolero PC. Video-assisted thoracic procedures: the
agement of lung cancer. Surgery alone, for many stages, mayo experience. Mayo Clin Proc. 1996;71:351.
is insufficient and the multidisciplinary approach to 14. Kadokura M, Colby TV, Myers JL, et al. Pathologic compar-
ison of video-assisted thoracic surgical lung biopsy with
therapy requires a coordinated approach by medical
traditional open lung biopsy. J Thorac Cardiovasc Surg.
oncology and radiation oncology. 1995;109:494.
15. Grogan EL, Jones DR, Kozower BD, Simmons WD,
Daniel TM. Identification of small lung nodules: technique
REFERENCES of radiotracer-guided thoracoscopic biopsy. Ann Thorac
1. Silvestri GA, Gonzalez AV, Jantz MA, et al. Methods for Surg. 2008;85(2):S772eS777.
staging non-small cell lung cancer: Diagnosis and manage- 16. Gelberg J, Grondin S, Trembley A. Mediastinal staging for
ment of lung cancer, 3rd ed: American College of Chest lung cancer. Can Respir J. 2014;21(3):159e161.
Physicians evidence-based clinical practice guidelines. 17. Yasafuku K, Pierre A, Darling G, et al. A Prospective
Chest. 2013;143(5):50Se211S. Controlled trial of Endobronchial Ultrasound-Guided Trans-
2. Deppen S, Putman JB, Andrade G, et al. Accuracy of FDG- bronchial Needle Aspiration Compared with Mediastinoscopy
PET to diagnose lung cancer in a region of endemic gran- for Mediastinal Lymph Node Staging of Lung Cancer. Philadel-
ulomatous disease. Ann Thorac Surg. 2011;92(2): phia, Pennsylvania: 91st Annual Meeting of The American
428e433. Association for Thoracic Surgery; 2011.
3. Khan KA, Nardelli P, Jaeger A, O’Shea C, Cantillon- 18. Darling GE, Dickie AJ, Malthaner RA, Kennedy EB, Tey R.
Murphy P, Kennedy MP. Navigational bronchoscopy for Invasive mediastinal staging of non-small-cell lung cancer:
early lung cancer: a road to therapy. Adv Ther. 2016; a clinical practice guideline. Curr Oncol. 2011;18(6):
33(4):580e596. e304ee310.
10 Pulmonary Adenocarcinoma: Approaches to Treatment

19. Wong RWM, Thai A, Khor YH, Ireland-Jenkin K, Lanteri CJ, 35. Gould G, Pearce A. Assessment of suitability for lung
Jennings BR. the utility of rapid on-Site evaluation on resection. Crit Care Pain. 2006;6(3):98e100.
endobronchial ultrasound guided transbronchial needle 36. Brunelli A, Belardinelli R, Refai M, et al. Peak oxygen con-
aspiration: does it make a difference? J Resp Med. 2014. sumption during cardiopulmonary exercise test improves
20. Kinsey CM, Arenberg DA. Endobronchial ultrasound- risk stratification in candidates to Major lung resection.
guided transbronchial needle aspiration for non-small Chest. 2009;135:1260e1267.
cell lung cancer staging. Am J Respir. 2015;189(6). 37. Lackey A, Donington JS. Surgical management of lung
21. Lee PC, Port JL, Korst RJ, Liss Y, Meherally DN, Altorki NK. cancer. Semin Intervent Radiol. 2013;30(2):133e140.
Risk factors for occult mediastinal metastases in clinical 38. Detterbeck FC, Boffa DJ, Kim AW, Tanoue LT. The eighth
stage I non-small cell lung cancer. Ann Thorac Surg. 2007; edition lung cancer stage classification. Chest. 2017;
84(1):177e181. 151(1):193e203.
22. Al-Sarraf N, Aziz R, Gately K, et al. Pattern and predictors 39. Donington JS, Pass HI. Surgical approach to locally
of occult mediastinal lymph node involvement in non- advanced non-small cell lung cancer. Cancer J. 2013;
small cell lung cancer patients with negative mediastinal 19(3):217e221.
update on positron emission tomography. Eur J Cardio- 40. Lanuti M. Surgical management of lung cancer involving
thorac Surg. 2008;33(1):104e109. the chest wall. Clinics. 2017;27(2):195e199.
23. Jiang L, Jiang S, Lin Y, et al. Nomogram to predict occult 41. Reardon ES, Schrump DS. Extended resections of non-
N2 lymph nodes metastases in patients with squamous small cel lung cancers invading the aorta, pulmonary ar-
non-small cell lung cancer. Medicine. 2015;94(46):e2054. tery, left atrium, or esophagus: can they be justified? Thorac
24. Defranchi SA, Cassivi SD, Nichols FC, et al. N2 Disease in Surg Clin. 2014;24(4):457e464.
T1 non-small cell lung cancer. Ann Thor Surg. 2009;88(3): 42. Van Schil PE, Berzenji L, Yogeswaran SK, Hendriks JM,
924e928. Lauwers P. Surgical management of stage IIIA non-small
25. Mason DP, Subramanian S, Nowicki ER, et al. Impact of cell lung cancer. Front Oncol. 2017;7:249.
smoking cessation before resection of lung cancer: a soci- 43. Farjah F, Wood DE, Varghese TK, Symons RG, Flum DR.
ety of thoracic surgeons general thoracic surgery database Trends in the operative management and outcomes of
study. Ann Thorac Surg. 2009;88(2):362e370. T4 lung cancer. Ann Thorac Surg. 2008;86(2):368e374.
26. Thomas DC, Blasberg JD, Arnold BN, et al. Validating the 44. David EA, Clark JM, Cooke DT, Melnikow J, Kelly K,
thoracic revised cardiac risk index following lung resection. Canter RJ. The role of thoracic surgery in the therapeutic
Ann Thorac Surg. 2017;104(2):389e394. management of metastatic non-small cell lung cancer.
27. Spyratos D, Zarogoulidis P, Porpodis K, Angelis N, et al. J Thorac Oncol. 2017;12(11):1636e1645.
Preoperative evaluation for lung cancer resection. J Thorac 45. Varela G, Thomas PA. Surgical management of advanced
Dis. 2014;6(supp1):S162e166. non-small cell lung cancer. J Thorac Dis. 2014;6(2):
28. Mazzone PJ. Preoperative evaluation of the lung cancer S217e223.
resection candidate. Expert Rev Respir Med. 2010;4(1): 46. Allen MS, Darling GE, Pechet TT, et al. Morbidity and mor-
97e113. tality of major pulmonary resections in patients with early-
29. Licker MJ, Widikker I, Robert J, et al. Operative mortality stage lung cancer: initial results of the randomized,
and respiratory complications after lung resection for can- prospective ACOSOC Z0030 trial. Ann Thorac Surg. 2006;
cer: impact of chronic obstructive pulmonary disease and 81(3):1013e1019.
time trends. Ann Thorac Surg. 2006;81:1830e1837. 47. Hazelrigg SR, Nunchuck SK, LoCicero J. Video assisted
30. Berry MF, Hanna J, Tong BC, et al. Risk factors for thoracic surgery study group data. Ann Thorac Surg. 1993;
morbidity after lobectomy for lung cancer in elderly 56:1039e1043.
patients. Ann Thorac Surg. 2009;88:1093e1099. 48. Roviaro G, Rebuffat C, Varoli F, Vergani C, Mariani C,
31. Brunelli A, Sabbatini A, Xiume F, et al. A model to predict Maciocco M. Videoendoscopic assisted pulmonary
the decline of the forced expiratory vlume in one second lobectomy for cancer. Surg Laparosc Endosc. 1992;2:
and the carbon monoxide lung diffision capacidty after 244e247.
major lung resection. Interact Cardiovasc Thorac Surg. 49. Abdelsattar ZM, Allen MS, Shen KR, et al. Variation in hos-
2005;4:61e65. pital adoption Rates of video-assisted thoracoscopic lobec-
32. Salati M, Brunelli A. Risk stratification in lung resection. tomy for lung cancer and the effects on outcomes. Ann
Curr Surg Rep. 2016;4(37):1e9. Thorac Surg. 2017;103(2):454e460.
33. Ferguson MK, Reeder LB, Mick R. Optimizing selection of 50. Blasberg JD, Seder CW, Leverson G, Shan Y, Maloney JD,
patients for major lung resection. J Thorac Cardiovasc Surg. Mackle RA. Video-assisted thoracoscopic lobectomy for
1995;109:275e281. lung cancer: current practice patterns and predictors of
34. Brunelli A, Refai MA, Salati M, Sabbatini A, Morgan- adoption. Ann Thorac Surg. 2016;102(6):1854e1862.
Hughes NJ, Rocco G. Carbon monoxide lung diffusion ca- 51. Boffa DJ, Allen MS, Grab JD, Gaissert HA, Harpole DH,
pacity improves risk stratification in patients without Wright CD. Data from the society of thoracic surgeons gen-
airflow limitation: evidence for systematic measurement eral thoracic surgery database: the surgical management of
before lung resection. Eur J Cardiothorac Surg. 2006;29: primary lung tumors. J Thorac Cardiovasc Surg. 2008;135:
567e570. 247e254.
CHAPTER 1 The Surgical Management of Pulmonary Adenocarcinoma 11

52. Flores RM, Ihekweazu U, Dycoco J. Video-assisted thoraco- 66. Park BJ, Melfi F, Mussi A, et al. Robotic lobectomy for non-
scopic surgery (VATS) lobectomy: catastrophic intraopera- small cell lung cancer (NSCLC): long-term oncologic
tive complications. Thorac Cardiovasc Surg. 2011;142: results. J Thorac Cardiovasc Surg. 2012;143(2):383e389.
1412e1417. 67. Reinersman JM, Passera E, Rocco G. Overview of uniportal
53. McKenna Jr RJ, Houck W, Fuller CB. Video-assisted video-assisted thoracic surgery (VATS): past and present.
thoracic surgery lobectomy: experience with 1,100 cases. Ann Cardiothorac Surg. 2016;5(2):112e117.
Ann Thorac Surg. 2006;81(2):421e425. 68. Song N, Zhao DP, Jiang L, et al. Suxiphoid uniportal video-
54. Onaitis MW, Petersen RP, Balderson S, et al. Thoracoscopic assisted thoracoscpoc surgery (VATS) for lobectomy: a
lobectomy is a safe and versatile procedure: experience report of 105 cases. J Thorac Dis. 2016;8(s3):251e257.
with 500 consecutive patients. Ann Surg. 2006;244(3): 69. Campione A, Ligabue T, Luzzi L, et al. Comparison be-
420e425. tween segmentectomy and larger resection of stage IA
55. Ali MK, Mountain CF, Ewer MS, Johnston D, Haynie TP. non-small cell lung carcinoma. J Cardiovasc Surg (Torino).
Predicting loss of pulmonary function after pulmonary 2004;45(1):67e70.
resection for bronchogenic carcinoma. Chest. 1980;77(3): 70. Martin-Ucar AE, Nakas A, Pilling JE, West KJ, Waller DA. A
337e342. case-matched study of anatomical segmentectomy versus
56. Rocco G, Internullo E, Cassivi SD, Van Raemdonck D, lobectomy for stage I lung cancer in high-risk patients.
Ferguson MK. The variability of practice in minimally inva- Eur J Cardiothorac Surg. 2005;27(4):675e679.
sive thoracic surgery for pulmonary resections. Thorac Surg 71. El-Sharif A, Gooding WE, Santos R. Outcomes of sublobar
Clin. 2008;18(3):235e247. resection versus lobectomy for stage I non-small cell lung can-
57. Demmy TL, Curtis J. Minimally invasive lobectomy cer: a 13-year analysis. Ann Thorac Surg. 2006;82:408e415.
directed toward frail and high-risk patients: a case-control 72. Kilic A, Schuchert MJ, Pettiford BL. Anatomic segmentec-
study. Ann Thorac Surg. 1999;68(1):194e200. tomy for stage I non-small cell lung cancer in the elderly.
58. Sugiura H, Morikawa T, Kaji M, Sasamura Y, Kondo S, Ann Thorac Surg. 2009;87:1662e1666.
Katoh H. Long-term benefits for the quality of life after 73. Koike T, Yamato Y, Yoshiya K. Intentional limited pulmo-
video-assisted thoracoscopic lobectomy in patients with nary resection for peripheral T1N0M0 small-sized lung
lung cancer. Surg Laparosc Endosc Percutan Tech. 1999; cancer. J Thorac Cardiovasc Surg. 2003;125:924e928.
9(6):403e408. 74. Iwasaki A, Hamanaka W, Hamada T, et al. Comparison be-
59. Petersen RP, Pham D, Burfeind WR, et al. Thoracoscopic tween a case-matched analysis of left upper lobe triseg-
lobectomy facilitates the delivery of chemotherapy after mentectomy and left upper lobectomy for small size
resection for lung cancer. Ann Thorac Surg. 2007;83(4): lung cancer located in the upper division. Thorac Cardiovasc
1245e1249. Surg. 2007;55(7):454e457.
60. Dylewski MR, Ohaeto AC, Pereira JF. Pulmonary resection 75. Cao C, Gupta S, Chandrakumar D, Tian DH, Black D,
using a total endoscopic robotic video-assisted approach. Yan TD. Meta-analysis of intentional sublobar resections
Semin Thorac Cardiovasc Surg. 2011;23(1):36e42. versus lobectomy for early stage non-small cell lung
61. Morgan JA, Ginsburg ME, Sonett JR, et al. Advanced thor- cancer. Ann Cardiothorac Surg. 2014;3:134e141.
acoscopic procedures are facilitated by computer-aided ro- 76. Okada M, Koike T, Higashiyama M, Yamato Y, Kodama K,
botic technology. Eur J Cardiothorac Surg. 2003;23: Tsubota N. Radical sublobar resection for small-sized non-
883e887. small cell lung cancer: a multicenter study. J Thorac Cardi-
62. Ashton Jr RC, Connery CP, Swistel DG, DeRose JJ. ovasc Surg. 2006;132:769e775.
Robot-assisted lobectomy. J Thorac Cardiovasc Surg. 77. Altorki NK, Yip R, Hanaoka T, et al. Sublobar resection is
2003;126:292e293. https://doi.org/10.1016/S0022-522 equivalent to lobectomy for clinical stage 1A lung cancer
3(03)00201-0. in solid nodules. J Thorac Cardiovasc Surg. 2014;147:
63. Gharagozloo F, Margolis M, Tempesta B. Robot-assisted 62e754; discussion 762e764.
thoracoscopic lobectomy for early-stage lung cancer. Ann 78. Blasberg JD, Harvey IP, Donington JS. Sublobar resection:
Thorac Surg. 2008;85(6):1880e1885. a movement from the lung cancer study group. J Thorac
64. Cerfolio RJ, Bryant AS, Skylizard L, Minnich DJ. Initial Oncol. 2010;5(10):1583e1593.
consecutive experience of completely portal robotic pul- 79. Guidoccio F, Orsini F, Mariani G. A critical reappraisal of
monary resection with 4 arms. J Thorac Cardiovasc Surg. sentinel lymph node biopsy for non-small cell lung
2011;142(4):740e746. cancer. Clin and Translational Imag. 2016;4(5):385e394.
65. Giulianotti PC, Buchs NC, Caravaglios G, Bianco FM. 80. Wang Y, Darling GE. Complete mediastinal lymph node
Robot-assisted lung resection: outcomes and technical dissection versus systematic lymph node sampling in sur-
details. Interact Cardiovasc Thorac Surg. 2010;11(4): gical treatment of non-small cell lung cancer: do we have
388e392. the answer? J Thorac Dis. 2017;9(11):4169e4170.
12 Pulmonary Adenocarcinoma: Approaches to Treatment

FURTHER READING 2. Darling GE, Li F, Patsios D, et al. Neoadjuvant chemoradia-


1. Cerfolio RJ, Maniscalco L, Bryant AS. The treatment of tion and surgery improves survival outcomes compared
patients with stage IIIA non-small cell lung cancer from with definitive chemoradiation in the treatment of stage
N2 disease: who returns to the surgical arena and who IIIA N2 non-small-cell lung cancer. Eur J Cardiothorac Surg.
survives. Ann Thorac Surg. 2008;86(3):912e920. 2015;48(5):684e690.
CHAPTER 2

Pulmonary
AdenocarcinomadPathology and
Molecular Testing
PRODIPTO PAL, MD, PHD • MICHAEL CABANERO, MD •
MING-SOUND TSAO, MD, FRCPC

INTRODUCTION as well as pulmonary blastoma. This chapter will focus


Lung cancer is the most common cause and one of the on pulmonary adenocarcinoma.
most aggressive human cancers in the world, with Adenocarcinomas are predominantly located in the
1.8 million new cases (13% of total cancer diagnoses) lung periphery.5 However, recent publications suggest
and 1.59 million deaths estimated to occur worldwide that central lung tumors often are histologically adeno-
in 2012.1,2 Pulmonary adenocarcinoma represents carcinomas; specifically 33% of solid predominant,
approximately 50% of all lung cancers and 60% of 25% of micropapillary predominant, and 20% of acinar
nonesmall cell carcinomas (NSCCs). In the last decade, predominant adenocarcinomas can be central tu-
more accurate histological diagnosis of adenocarci- mors.6,7 On gross examination, most adenocarcinomas
noma has become essential in selecting patients for appear as irregular-shaped tan-gray to gray-white nod-
testing of molecular alterations with effective targeted ules and are often associated with scarring, anthracotic
therapies. This importance is recognized in the 2015 pigment deposition, and pleural puckering if close to
World Health Organization (WHO) classification of the pleura.8 The so-called noninvasive tumors with lepi-
lung tumors, with subtype-specific immunomarkers be- dic growth of tumor cells on preexisting alveolar archi-
ing integrated into the diagnostic criteria of nonesmall tecture are often difficult to identify on fresh specimens.
cell lung cancers (NSCLCs), especially for poorly differ- Major changes to the classification of lung adenocar-
entiated tumors in small biopsy samples.2,3 cinoma were proposed in 2011 by the International As-
sociation for the Study of Lung Cancer (IASLC),
American Thoracic Society (ATS), and European Respi-
HISTOLOGICAL CLASSIFICATION ratory Society (ERS).9 For the first time, this proposal
The first attempt at morphological classification of pul- recognized lung adenocarcinoma with pure lepidic
monary carcinomas was proposed by Marchesani in growth pattern as adenocarcinoma in situ (AIS), and a
1924 and was later incorporated in the WHO histolog- predominantly AIS tumor with <0.5 cm area of inva-
ical classification of lung tumors of 1967.4 Since then, sion as minimally invasive adenocarcinoma (MIA).
the classification of pulmonary tumors has undergone AIS and MIA are uncommon, encompassing only 5%
multiple iterations with significant updates, especially of resected adenocarcinomas.
for adenocarcinoma. The major subtypes in the 2015 Pulmonary adenocarcinoma is further subclassified
WHO classification include adenocarcinoma, squa- according to cell type into nonmucinous and mucinous
mous cell carcinoma, neuroendocrine tumors which adenocarcinoma subtypes. A more detailed histological
include small cell carcinoma and large cell neuroendo- subtyping is rendered in resection specimen. The most
crine carcinoma, and large cell carcinoma (Table 2.1). common invasive adenocarcinoma is classified based
The relatively rarer subtypes include salivary gland on five histological growth patterns: lepidic, acinar,
type carcinoma (e.g., mucoepidermoid, adenoid cystic) papillary, micropapillary, or solid (Fig. 2.1). As most
and sarcomatoid carcinoma, which encompass carcino- adenocarcinomas demonstrate complex and heteroge-
sarcoma, pleomorphic/spindle cell, giant cell carcinomas, neous growth patterns, it has been proposed that the

Pulmonary Adenocarcinoma: Approaches to Treatment. https://doi.org/10.1016/B978-0-323-55433-6.00002-X


Copyright © 2019 Elsevier Inc. All rights reserved. 13
14 Pulmonary Adenocarcinoma: Approaches to Treatment

TABLE 2.1
Major Classification of Pulmonary Nonesmall Cell Carcinomaa
Major Histological Type Subtype Diagnostic Histological Features
Squamous cell carcinoma In situ carcinoma Full thickness dysplastic change and mitotic figures in
bronchial/bronchiolar epithelium
Keratinizing Squamous “keratin pearls,” intercellular bridges
IHC: p40/p63þ (diffuse)
Nonkeratinizing No morphological evidence of squamous differentiation
IHC: p40/p63þ
Basaloid Monotonous hyperchromatic tumor cells with palisading;
“comedo-type” necrosis common
IHC: p40/p63þ
Adenocarcinoma
Nonmucinous adenocarcinoma Lepidic Tumor cell growth along preexisting alveolar septa
Pure lepidic growth is considered in situ adenocarcinoma
Acinar Gland formation with central lumina and lined by tumor cells
Papillary Glandular cells with fibrovascular core
Micropapillary Tufts of cells lacking fibrovascular cores and often detached
Solid Sheets of polygonal cells lacking overt glandular pattern
Tumor cells stain positive for TTF-1 by
immunohistochemistry or demonstrate presence of
intratumoral mucin
Invasive mucinous Columnar or goblet cells with abundant intracytoplasmic
adenocarcinoma mucin Can be TTF-1 negative (85%)
Other variants Colloid Alveolar space filled with mucin pools that destroy alveolar
wall structures
Fetal Complex glands with nonciliated glycogen-rich tumor
resembling fetal lung
Enteric Resemble colorectal adenocarcinoma with complex glands
formed by tall columnar cells, resembling intestinal cells
Adenosquamous carcinomaa Both adenocarcinoma and squamous cell carcinoma
components are present
At least 10% of any component required
Large cell carcinomaa Undifferentiated nonesmall cell carcinoma
Diagnosis of exclusion and should be reserved for resection
specimens
Sarcomatoid carcinomaa Pleomorphic Poorly differentiated NSCC with at least 10% spindle or giant
cells
Spindle cell Purely composed of spindle cells
Giant cell Purely composed of giant cells
Carcinosarcoma Biphasic tumor with nonesmall cell carcinoma component
(adenocarcinoma or squamous cell carcinoma) and
sarcomatous heterologous elements
Pulmonary Biphasic tumor with primitive mesenchymal stroma and fetal
blastoma adenocarcinoma
a
Adapted from the 2015 WHO Classification.1
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 15

A B

C D

E F
FIG. 2.1 Subtypes of pulmonary adenocarcinoma. (A) Lepidic pattern; (B) Acinar pattern in a biopsy
specimen and focal micropapillary features (bottom right); (C) Papillary pattern; (D) Micropapillary pattern;
(E) Solid pattern and corresponding TTF-1 IHC (F). Note: (AeE): Hematoxylin and eosin stains.

breakdown of histological subtypes should be esti- specimens. For most diagnostic specimens, which are
mated in 5% increments, and the tumor is classified ac- small biopsy or cytology specimens, subtype classifica-
cording to its most common or predominant pattern. tion is not necessary, but there are recommended guide-
This subtype classification requires the examination of lines to use standardized nomenclature for diagnosis.1
the entire tumor, thus is mainly applicable to resection The accepted terminologies include NSCC, favor
16 Pulmonary Adenocarcinoma: Approaches to Treatment

adenocarcinoma, squamous cell carcinoma, or not currently classified under acinar histological pattern. It
otherwise specified, depending on morphological char- is important to mention cribriform architecture as it
acteristics or immunohistochemical (IHC) marker has been reportedly associated with poor prognosis12
expression. The commonly used IHC markers for and often associated with the gene-rearranged tumors
adenocarcinoma are pneumocyte markers such as thy- such as ALK, ROS1.13 Papillary pattern shows growth
roid transcription factor 1 (TTF-1), napsin A, and CK7; of neoplastic cells along a central fibrovascular core.
for squamous cell carcinoma, they include p40 (or Papillary pattern corresponds to invasive tumor compo-
p63) and/or CK5/6. With a combination of 2e3 nent, even in the absence of myofibroblastic stroma.
markers (TTF-1, p40/p63, and CK5/6), more than Across multiple studies, acinar and papillary predomi-
90% of poorly differentiated NSCLC biopsy specimens nant tumors have been regarded as intermediate risk
can be classified as favoring one of the subtypes. In with a 5-year DFS of w80%e85%.14,15
addition to the above subtypes, several less common
variants include invasive mucinous adenocarcinoma Micropapillary and Solid Predominant
(IMA), colloid adenocarcinoma, enteric adenocarci- Adenocarcinoma (Poor Prognosis)
noma, and fetal adenocarcinoma. Both micropapillary and solid tumors are considered
high-grade adenocarcinoma with poor prognoses. This
prognostic association has been reported across multi-
CLINICAL RELEVANCE OF ple series. Micropapillary tumor commonly appears de-
ADENOCARCINOMA SUBTYPE tached; grows in papillary tufts devoid of fibrovascular
CLASSIFICATION cores; and has a high rate of lymphovascular invasion,
Lepidic Predominant Adenocarcinoma STAS, and recurrence.14-17 In contrast, solid pattern tu-
(Good Prognosis) mor is composed of sheets of polygonal cells and lack
In the most recent 2015 WHO tumor classification, the morphologic evidence of glandular architecture. The
term “lepidic” refers to noninvasive tumor. Tumors solid pattern adenocarcinoma shows similar aggressive
such as AIS and MIA are both lepidic tumors and differ behavior as micropapillary tumors.14e16,18
primarily on the presence or absence of focal invasion.
Invasive tumor is defined as any histology other than Invasive Mucinous Adenocarcinoma
lepidic pattern (e.g., acinar, papillary, micropapillary, The tumor cells have columnar or goblet cell
and solid) and the presence of myofibroblastic stromal morphology with basally oriented nuclei and abundant
reaction in association with tumor cell invasion, intracytoplasmic mucin. IMA has a strong tendency for
vascular, or pleural invasion, and spread through air multicentric bilateral tumor at presentation.1,2 All the
spaces (STAS). Lepidic growth can also be seen in above architectural excluding solid pattern can be seen
IMAs, but the latter invariably also include the presence in IMA. Often times, metastatic mucinous tumors
of other growth patterns. However, “lepidic” predomi- from pancreas, lower gastrointestinal tract, or ovary
nant adenocarcinoma (LPA) essentially refers to non- appear morphologically identical and can be difficult
mucinous adenocarcinoma with predominantly to distinguish. Pulmonary IMA frequently show muta-
lepidic but also demonstrate invasive patterns. tions in KRAS (up to 90%)19e22 and NRG1 fusions.23,24
The distinction between AIS and MIA is primarily on
the absence or presence of invasive focus; MIA by defi-
nition cannot have an invasive focus more than PATHOLOGICAL STAGING OF LUNG
0.5 cm and features such as lymphovascular or pleural ADENOCARCINOMAdUICC/AJCC EIGHTH
invasion, STAS, or tumor necrosis.1 AIS and MIA have EDITION
close to 100% 5-year disease-free survival (DFS).10,11 The new (eighth) edition of the lung cancer staging clas-
LPA accounts for tumors with >0.5 cm invasive foci. A sification system developed by the International Associ-
large number of publications have demonstrated that ation for the Study of Lung Cancer (IASLC) Staging and
lepidic predominant adenocarcinoma carries a good Prognostic Factor Committee has been approved by
prognosis for the patients. both Union for International Cancer Control (UICC)
and American Joint Committee on Cancer (AJCC).25e29
Acinar/Papillary Predominant While UICC activates this new system in 2017, the
Adenocarcinoma (Intermediate Prognosis) implementation by AJCC is delayed until January 1,
Acinar pattern morphologically refers to round- to oval- 2018.
shaped glands with central lumina. Cribriform architec- The main changes in the AJCC eighth edition corre-
ture, where tumor glands are often fused together, is sponds to T-descriptor or tumor size and extent and
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 17

changes in M-descriptor; there is no formal change in tumors with diaphragm and recurrent laryngeal
the N-staging. As noted earlier, the in situ carcinoma nerve involvement have now been updated to pT4
(AIS or squamous in situ carcinoma [SCIS]) and MIA tumors.
have been formally introduced as Tis in the AJCC eighth In the M-category, the definition of M1a (separate
edition of TNM classification. The T-descriptor changes tumor nodule(s) in a contralateral lobe; tumor with
correspond to (1) tumor size, (2) staging of multiple/ pleural or pericardial nodule(s), or malignant pleural
synchronous lung tumors, and (3) updating tumor or pericardial effusion) is not changed. However, the
spread/extent. The early-stage tumors (previously pT1a M1b category has been expanded to distinguish be-
and pT1b in AJCC seventh edition) have been expanded tween extrathoracic oligometastatic (M1b) and tumors
to three subcategories in the AJCC eighth edition, i.e., with multiple metastatic disease (M1c).
pT1a (1 cm), pT1b (>1e2 cm), and pT1c
(>2e3 cm). The size criteria for prior pT2 and pT3 tu-
mors from AJCC seventh edition now have been MOLECULAR CLASSIFICATION
updated. A lung tumor of >3e5 cm now corresponds The recent coordinated effort by The Cancer Genome
to pT2 tumor (pT2a: >3e4 cm, pT2b: >4e5 cm), while Atlas (TCGA) to classify recurrent molecular alterations
pT3 tumors are >5e7 cm. Any tumor >7 cm is now in multiple cancers has revealed 18 statistically signifi-
classified as pT4. cant altered genes in lung adenocarcinoma.35 These
In the new eighth edition of the UICC/AJCC tumor include TP53 (46%), KRAS (33%), EGFR (14%), BRAF
staging classification, the pathological T stage is also (10%), PIK3CA (7%), MET (7%), RIT1 (2%), STK11
based on the size of the “invasive,” nonlepidic histolog- (17%), KEAP1 (17%), NF1 (11%), RB1 (4%), CDKN2A
ical component of the adenocarcinoma.30 This has sig- (4%), SETD2 (9%), ARID1A (7%), MARCA4 (6%),
nificant implication, as semiquantitative estimation of RBM10 (8%), U2AF1 (3%), and MGA (8%). Recur-
the “noninvasive” lepidic component and morphologic rently altered/activated cellular pathways were also
distinction of other invasive patterns are of utmost identified and include activation of RTK/RAS/RAF
importance. pathway, the PI(3)K-mTOR pathway, p53 pathway,
The concept of intrapulmonary metastasis has not cell-cycle regulator pathway, oxidative stress pathways,
been changed: ipsilateral same-lobe tumor nodule and mutations in chromatin and RNA splicing factors.
(pT3), ipsilateral different-lobe tumor nodule (pT4), Molecular classification from the perspective of target-
and contralateral tumor nodule (M1a). While the stag- able oncogene drivers and the various methods for
ing of multifocal lung cancers has been ambiguous in detection will be reviewed later.
the AJCC seventh edition, this has been addressed in a
series of new publications.31e33 The critical point is to
distinguish synchronous separate primary lung tumors TRANSCRIPTOMIC CLASSIFICATIONS
and metastatic tumors. Various criteria have been pub- Although gene expression profiles have been used to
lished to distinguish synchronous versus metastatic tu- subclassify a tumor’s molecular phenotype and
mor. Briefly, if the tumor differs according to cell type behavior into clinically relevant groups,36 routine
(e.g., adenocarcinoma vs. squamous cell carcinoma) profiling of a tumor’s transcriptome has not reached
or by comprehensive histological patterns, a synchro- standard clinical practice. Nevertheless, we include a
nous but separate primary tumors are favored. While brief discussion on gene expression subclassification
separate primaries should be individually staged, the to help shed insight into the biology of these tumors.
staging of multiple lung cancers with multiple Over the years, multiple studies have observed three
ground-glass opacities on imaging studies shows reproducible distinct subtypes of lung adenocarcinoma
low-grade morphology in the lepidic predominant based on the tumor’s transcriptome.35e40 They were
spectrum on pathology (AIS or MIA) is now staged designated with various names, such as bronchioid,
with the tumor with highest stage with the letter “m” squamoid, and magnoid, as they appeared to have
in parenthesis.32e34 Pneumonic type adenocarcinoma, similar expression profiles as bronchioalveolar carci-
most commonly seen in IMA, is staged based on the noma (now-termed lepidic adenocarcinoma), squa-
size of the tumor and location (pT3 in single lobe, mous cell carcinoma, and large cell carcinoma,
pT4 involving different ipsilateral lobe or M1a respectively.38 However, these subtype nomenclatures
involving contralateral lobes). In the eighth edition, were revised in the TCGA manuscript to synergize the
tumors involving main bronchus regardless of distance different transcriptional subtypes with tumor histology,
from carina are now staged as pT2 tumors, while anatomic locations, and mutational profile.35
18 Pulmonary Adenocarcinoma: Approaches to Treatment

Terminal Respiratory Unit Subtype Peutz-Jeghers syndrome,43,44 and studies have found
The terminal respiratory unit (TRU), formerly known as sporadic STK11 mutations in lung
bronchioid, is composed of lung adenocarcinomas that adenocarcinomas.45e47 Similar to patients with PI
have cell lineage properties linking them to respiratory tumors, patients with PP tumors had worse survival
epithelium of the TRU.41 Tumors in the TRU transcrip- than those with TRU tumors.35,38,39
tional subunit account for approximately 30% of lung
adenocarcinomas38 and have the highest proportion
of tumors that express TTF-1. TRU tumors also maintain ONCOGENIC DRIVER MUTATIONS AND
morphologic similarity to the benign cells of the TRU. MOLECULAR TESTING IN LUNG
Under the current WHO classification,3 most nonmuci- ADENOCARCINOMA
nous adenocarcinomas with lepidic and acinar patterns, Molecular Testing Methods
as well as some papillary adenocarcinomas, belong to The various assays used to detect recurrent molecular al-
this group. Compared with the other two groups, tu- terations in NSCLC, including epidermal growth factor re-
mors of the TRU transcriptional subtype have the best ceptor (EGFR) mutations and structural alterations
clinical outcomes in early-stage disease,35,38,39 which involving the anaplastic lymphoma kinase (ALK) and
may be explained by its overall lower rate of distant me- ROS1 genes, have evolved over the past decade. Starting
tastases in early stage. The majority of EGFR-mutant tu- with single-gene sequencing (i.e., Sanger sequencing) or
mors, and those harboring kinase-fusion alterations, fluorescence in situ hybridization (FISH), many newer
belong to the TRU,38 with increased prevalence of tests use massive parallel sequencing technology, or
younger patients, female gender, and nonsmokers.41 NGS to target multiple verified and potentially “drug-
gable” genes implicated in oncogenesis. Broadly, these
Proximal Inflammatory Subtype techniques can be categorized into either nontargeted
The proximal inflammatory (PI) unit, formerly known (detect new or known mutations) or targeted (detect
as squamoid, is enriched for high-grade tumors with only known mutations) assays, and whether single
solid histomorphology and accounts for approximately gene/locus (uniplexed) or multiple genes/loci (multi-
50% of lung adenocarcinomas.38 Patients tended to be plexed) are interrogated (Table 2.2).
male smokers with tumors that metastasized early and
had a higher proportion of central nervous system
involvement. In comparison to the TRU subtype, pa- EPIDERMAL GROWTH FACTOR RECEPTOR
tients with PI subtype tumors had poorer prognosis in The EGFR (also known as HER1 or ERBB1) gene,
early-stage disease but fared better in late-stage disease. located on chromosome 7, is a transmembrane protein
Histologically, PI tumors tended to be moderately to and a member of the epidermal growth factor (EGF)
poorly differentiated, had a high degree of TP53 and family. The EGF family includes four members (HER1
NF1 tumor suppressor mutations, and a large degree through HER4) and functions to bind extracellular pro-
of p16 inactivation.40 Pathways activated in these tu- tein ligands to activate an array of cellular pathways that
mors include WNT signaling, TGF-b signaling, and include differentiation, proliferation, and survival of
angiogenesis (HIF1a). the cell.48 Ligand-receptor interaction leads to dimeriza-
tion of the EGFR with either itself (homodimerization)
Proximal Proliferative Subtype or with another member of the EGFR family proteins,
The proximal proliferative (PP) unit, formerly known as i.e., HER2, HER3, and HER4 (heterodimerization).
magnoid, accounts for approximately 20% of lung ade- Downstream pathways activated include the RAS-RAF-
nocarcinomas. This subtype was originally termed mag- MAPK and the PI3K-AKT-mTOR signaling cascade. Acti-
noid as genes expressed in this subtype were most vation of EGFR ultimately leads to transcription of
similar to those found in large cell carcinomas.38,40 genes that promote cell-cycle progression, proliferation,
These tumors are particularly enriched for KRAS muta- and survival.
tions. They also included a high degree of STK11 (aka Mutations in the EGFR account for approximately
LKB1) inactivation, via chromosomal loss, inactivating 15% in Western countries and up to 50% in Asian coun-
mutations, or reduced gene expression.35 STK11 is a tu- tries of lung adenocarcinoma cases, with a higher pro-
mor suppressor that regulates growth and metabolism portion found in younger age, East Asian, women,
when nutrients are scarce by acting upstream to adeno- and light/never smokers.49,50 Almost all sensitizing mu-
sine monophosphate kinase (AMPK).42 Germline tations (activating mutations upon which TKI therapy
mutations in the STK11 gene are associated with has shown efficacy) occur between exons 18e21. The
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 19

(2) activation of bypass pathways, (3) activation of down-


TABLE 2.2
stream pathways, and (4) histologic transformation.
Current Actionable Target Genes in Lung
Adenocarcinoma and Methods for Detection Secondary Resistance Mutations
Target The most common resistance mechanism is the T790M
Gene Assay Examples mutation in exon 20 (kinase domain), and it accounts
for approximately 60% of all acquired resistance to
EGFR Nontargeted
assaysdSanger EGFR-TKI therapy. Arising in cis with the sensitizing
sequencing, EGFR mutation, this “gatekeeper” mutation restores
pyrosequencing the binding pocket’s affinity for adenosine triphosphate
(ATP).51 This effectively renders the competitive inhibi-
Targeted Cobas (Roche,
assaysdallele- Tucson, AZ), tion of first- and second-generation TKIs ineffective to
specific real-time Therascreen ATP.52,53 Other rare kinase domain secondary EGFR
PCR, PCR (Qiagen, Hilden, mutations have been associated with TKI resistance,
amplification with Germany), including L747S, D761Y, and T854A.54,55 In 10% of pa-
allele-specific base MassARRAY (Agena tients with T790M mutations, there is a concomitant
extension Bioscience, San amplification of EGFR and is thought to outcompete
Diego, CA) second-generation TKI inhibition by providing stronger
ALK/ Structural kinase domain activation.56
ROS1 alterationsdFISH
Activation of Bypass Pathways
Multiple Multiplex RT-PCR SNaPshot Kit
genes (Applied MET amplification accounts for 5% of resistance
Biosystems, Foster mechanisms; this amplification allows for excess MET
City, CA) receptors to heterodimerize with HER3 to activate
Next-generation Oncomine PI(3)K-Akt-mTor signaling despite TKI inhibition.57
sequencing panels (Thermofisher, Increased expression of hepatocyte growth factor, the
Waltham, MA), ligand for MET, can also occur.58 Finally, HER2
FoundationOne (ErbB2) is found in approximately 8%e13% of those
(Foundation with EGFR-sensitizing mutations and leads to parallel
Medicine, MA), signaling of downstream pathways that bypass the ef-
TruSeq Amplicon fects of EGFR-TKIs.59
(Illumina, San Diego,
CA) Activation of Downstream Pathways
Whole exome/ BRAF mutations (1%) and MAPK1 amplifications
genome sequencing (<1%) drives continued signaling of the RAS/RAF/
MAPK pathway.60 Similarly, PTEN decrease or loss
and PIK3CA mutations (5%) activate the PI3K-AKT-
mTOR pathway even in the presence of EGFR-TKI.61

most common mutations include a short in-frame dele- Histologic Transformation


tion at exon 19, which primarily affects amino acid res- Accounting for 5%e14% of acquired resistance mecha-
idues 747e750, as well as the missense mutation nisms, small cell lung carcinoma (SCLC) transforma-
L858R in exon 21. These two mutations account for tion has been observed since the early days of EGFR
approximately 85%e90% of all EGFR lung adenocarci- targeted therapy.62 The mechanism for resistance seems
noma mutations, but many other mutations outside to be correlated with decreased EGFR expression in
these hotspots have shown TKI response. SCLC.63 Epithelial to mesenchymal transition (EMT)
Despite improved clinical response compared to is characterized by histologic change of the epithelial tu-
chemotherapy, acquired resistance to TKI therapy mor cells to spindle cell morphology and a molecular
commonly occurs in patients with EGFR-sensitizing change of decreased expression of the epithelial marker
mutations. These can be broadly classified into four cat- E-cadherin and increased expression of the mesen-
egories: (1) secondary resistance EGFR mutations, chymal marker Vimentin.64
20 Pulmonary Adenocarcinoma: Approaches to Treatment

Epidermal Growth Factor Receptor Mutation amplification-refractory mutation system (ARMS).


Testing Therascreen is able to identify 29 EGFR mutations and
Nontargeted assays was the companion diagnostic in the LUX-Lung 3 trial
Sanger sequencing was the standard for EGFR testing in that led to the approval of afatinib.
the first clinical trials with erlotinib and gefitinib. This
method incorporates fluorescent-tagged dideoxy termi-
nators to amplifying DNA strands, which can be sorted ANAPLASTIC LYMPHOMA KINASE
by size and the nucleotide sequence read sequentially. A ALK, located on chromosome 2p23, encodes a receptor
major consideration is the relatively low analytical tyrosine kinase (RTK) and was first described in
sensitivity of the assay, which usually requires speci- anaplastic large cell lymphoma.65 ALK rearrangements
mens with high tumor content. As such, this assay is were reported in pulmonary adenocarcinoma in
no longer a method of choice for detection of EGFR mu- 2007.66,67 The most common fusion partner is EML4;
tations. Another method, called pyrosequencing, in- however, over 20 different ALK gene partners have been
volves measuring the chemiluminescent signal described in recent times.68e71 The breakpoints on the
released by pyrophosphate as triphosphate nucleotides ALK gene most commonly happen at intron 19, or rarely
are being incorporated into the synthesized DNA at exon 20, but preservation of the kinase domain in the
strand. Although the fragment length required for pyro- resulting fusion gene occurs universally. Typically, the
sequencing is much shorter than those used for Sanger fusion partners contribute coil-coil domains which al-
sequencing, this method offers higher sensitivity and lows spontaneous dimerization of ALK fusion proteins
can detect mutations in samples with up to 5% tumor and subsequently results in constitutive activation of
cellularity. the oncogenic fusion gene products. ALK-rearranged pul-
monary adenocarcinoma has gained widespread popu-
Targeted assays larity again due to the availability of small molecule
Targeted assays, often used to detect specific EGFR mu- TKIs. Several tyrosine kinase inhibitors are now available
tations, offer better sensitivity than nontargeted for the treatment of ALK-rearranged lung tumors.
sequencing assays. Most current platforms require The estimated frequency of NSCLC harboring ALK
only approximately 5%e10% tumor cellularity and rearrangement is 3%e5%.72 ALK-rearranged tumors
can interrogate multiple hotspot mutations in a single are associated with female sex, younger patients, and
test. The USDA-approved companion diagnostic assay those with never to light-smoking history.73e75 As
for erlotinib, the Cobas EGFR Mutation Test (Roche), with EGFR, ALK rearrangements are found in adenocar-
uses allele-specific primers to identify the most com- cinomas and are uncommon in squamous histology or
mon EGFR mutations, using both tissue and plasma neuroendocrine carcinomas. Pulmonary adenocarci-
samples, and can interrogate 42 clinically relevant mu- noma with mucinous features, acinar/cribriform archi-
tations, including T790M. The Therascreen EGFR RCQ tecture, signet ring cells, and psammomatous
PCR kit (Qiagen) is another platform that uses allele- calcification are the histologies associated with ALK-
specific primers, in the form of Scorpion primers, or rearranged tumors13 (see Fig. 2.2). Thus, both EGFR

A B
FIG. 2.2 ALK-rearranged pulmonary adenocarcinoma. (A) The tumor is arranged in acinar/cribriform
architecture, with mucinous features; signet ring morphology is present in lower half (hematoxylin and eosin
stain); (B) IHC for ALK (5A4 clone) shows diffuse cytoplasmic, often granular ALK expression in tumor cells.
ALK, anaplastic lymphoma kinase; IHC, immunohistochemical.
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 21

and ALK testing mostly are reserved for patients with fusion variants such as SLC34A2-ROS1, TPM3-ROS1,
adenocarcinoma histology and not recommended for SDC4-ROS1, EZR-ROS1, and others. Similar to ALK,
pure squamous cell or neuroendocrine carcinomas.76 ROS1 fusion tumors are enriched in females, never
It should be noted, however, that ALK rearrangement smokers,88 and with adenocarcinoma histology.86,89,90
as well as ALK protein expression by IHC rarely has Cribriform/acinar architecture, mucinous features, solid
been reported in squamous cell carcinoma,77,78 pattern histology, hepatoid morphology, signet ring cell
although it has been suggested that this finding may morphology, and tumors with psammomatous calcifi-
be related to existing challenges in diagnosing NSCLC cation are the most common histologic patterns
in small biopsies and limitation of representative tumor reported in ROS1 fusion pulmonary adenocarci-
sampling. noma.91,92 ROS1 fusions has not been described in
Much like the case with TKI-treated patients with squamous cell carcinoma or lung neuroendocrine
EGFR-sensitizing mutations, patients with ALK rear- carcinoma.
rangements treated with crizotinib almost always The interest in ROS1 fusions stems from the avail-
develop secondary acquired resistance mutations. These ability of targetable small molecular TKI, crizotinib.93
mutations lead to a decrease in binding affinity for cri- The published clinical trials corroborate significant clin-
zotinib or an increased binding affinity to ATP.79 Other ical activity of crizotinib in ROS1 fusion tumors.89,90
resistance mechanisms have also been described and Following the clinical trials, multiple acquired resis-
include development of secondary EGFR or KRAS muta- tance mutations have also been reported.94,95 Fortu-
tions, and ALK and KIT gene amplifications.80e82 nately, the second-generation TKIs were developed
within short time that were able to target most of the
ROS1 resistance mutations.96e98
ROS1 is an RTK of the insulin receptor family, located
on chromosome 6q22 and encodes a transmembrane
protein with intracellular C-terminal tyrosine kinase ALK AND ROS1 TESTING
domain and shares marked sequence homology and As a general rule, proper handling and processing of tis-
structural similarities to ALK oncogene.83 ROS1 fusion sue, adequacy of diagnostic material are quintessential
was first described in glioblastoma multiforme,84,85 steps in assessing and testing for biomarker studies.99
later reported in NSCLC by Rikova et al.67 The first pop- Tissue fixation with 10% neutral buffered formalin
ulation screening of ROS1 in NSCLC was reported by and tissue fixation of 6e72 h are recommended.72
Bergethon et al.86 Subsequently, over 20 ROS1 fusions The main methods to detect both ALK and ROS1 rear-
have been described in NSCLC as well as in other solid rangements are (1) FISH, (2) real-time polymerase
tumors. The precise mechanism of oncogenic activity chain reaction or RT-PCR, (3) IHC, and (4) NGS.
and constitutive expression of ROS1 fusion products
are as yet unclear. Break Apart FISH Assay
The prevalence of ROS1 fusion in NSCLC is esti- ALK and ROS1 rearrangement detection by FISH is
mated to be w1%e2%.72,86,87 CD74-ROS1 fusion is achieved by using dual-colored break apart probes
the most common fusion variant,87 followed by other that label the 3ʹ (telomeric) and 5ʹ (centromeric) part.

A B
FIG. 2.3 (A) PD-L1 (SP263 clone) IHC is evaluated as partial or complete tumor membranous staining.
(B) Many nonneoplastic cells may also express PD-L1, including lymphocytes (shown here), macrophages,
and endothelial cells. Care must be taken to distinguish the cells from tumor cells for appropriate scoring.
PD-L1, program death ligand 1.
22 Pulmonary Adenocarcinoma: Approaches to Treatment

The ALK Vysis LSI break apart FISH probe (Abbott Mo- 5A4 (Novocastra), D5F3 (Cell Signaling Technology),
lecular) was approved by Food and Drug Administra- and anti-ALK (Origene). ALK1 antibody (Dako) is less
tion (FDA) for the therapy with ALK inhibitors and is sensitive and should not be used. The other clones, spe-
commonly used worldwide (US Food and Drug Admin- cifically D5F3 (as companion diagnostic assay) and 5A4
istration). In contrast, there are several break apart FISH (laboratory-developed test), have been widely studied
probes for ROS1, but none has been FDA approved as and have shown to be equally sensitive across multiple
companion diagnostics. A minimum of 50 tumor cells large-scale studies.72 ALK staining is cytoplasmic and
are required and a cutoff of 15% is the general recom- granular in character and can occasionally demonstrate
mended guideline. Inaccurate signal interpretation, membrane accentuation (Fig. 2.2). It is important to be
borderline rates of rearrangement positive cells, erro- cognizant of various artifacts such as evaluating ALK
neous results due to false-positive or false-negative re- staining in mucin-producing cells (occasionally in
sults, and atypical signal profile are the challenges in extracellular mucin, as well as cytoplasm of alveolar
test interpretation errors.100 macrophages and bronchial cells) and nonspecific
membranous staining and staining in squamous cell
Reverse Transcription-Polymerase Chain carcinoma (rare instances), large cell neuroendocrine
Reaction carcinomas, and normal ganglion cells. The reproduc-
Reverse transcription-polymerase chain reaction ibility of ALK IHC results across different laboratories
(RT-PCR) is feasible in clinical laboratories, however, and pathologists is high for standardized/validated
with its own set of challenges. As noted earlier, ALK rear- procedures.108
rangement has many different candidate fusion part- IHC detection approach for ROS1 and antibody
ners, even for the ALK-EML4, the most common clone D4D6 (Cell Signaling Technology, MA) was intro-
fusion in NSCLC, there are many fusion variants, largely duced by Rimkunas et al.109 Multiple studies have
due to different breakpoint regions on EML4, thus shown adequate sensitivity of IHC for detecting ROS1
would require a multiplexed approach. However, with rearrangements; however, the key problem remains
RT-PCR-based methods, a priori knowledge of the with the specificity of the assay.87,110 Although there
candidate fusion genes is needed to identify fusion var- have been several publications on ROS1 IHC, the stain-
iants which can later be confirmed by subsequent ing protocol is yet to be standardized. Detailed discus-
sequencing101 and has been used in studies with high sion on ROS1 IHC can be found in the IASLC Atlas of
level of sensitivity albeit with varying level of specificity ALK and ROS1 Testing in Lung Cancer.72
(85%e100%) compared with FISH.102,103 A negative
result by RT-PCR requires appropriate caution due to Next-Generation Sequencing
the potential of false-negative results due to the missing NGS offers the best single platform option to simulta-
unknown fusion variants. In addition, RT-PCR assays neously detect driver mutations (e.g., mutations in
are dependent on procuring high-quality RNA from EGFR, KRAS, etc.), fusion-type structural variants
FFPE tissue. Newer RNA-based assays are under active (ALK, ROS1, RET, NTRK1, etc.), gene amplification
development (assays such as NanoString system) that (e.g., MET), or deletions. A number of NGS-based ap-
can simultaneously detect various fusion partners as proaches have been described recently including multi-
well as offer the added advantage of detecting other plexed PCR-based gene panels, hybrid captureebased
driver fusion-type alterations in a multiplexed setup gene panels, and comprehensive genome/exome
(such as ROS1, RET, etc.).104e106 sequencing.111e115
Specifically, in a large study of NSCLC, 4.4% (47/
Fusion Protein Detection of ALK and ROS1 1070 cases) ALK fusions were detected by NGS
by IHC approach; however, of these cases, FISH data were avail-
ALK protein expression in NSCLC is generally lower in able for a subset which revealed a significant percentage
comparison with ALK expression in anaplastic large of cases were reported as FISH negative (35%).116 Mul-
cell lymphoma.107 Therefore, it is important to stan- tiple large-scale and independent studies now have
dardize ALK IHC assay and adequately address various established higher sensitivity/specificity for detection
issues pertaining to antigen retrieval, higher affinity pri- of actionable genomic alterations with NGS-based
mary antibody in higher concentration, and stronger approaches.111,114,116,117
signal amplification steps.72 There are four commer- The limitations of NGS, especially in a clinical labo-
cially available antibodies to date, viz., ALK1 (Dako), ratory setting include cost, high-level complex
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 23

laboratory setup with in-depth bioinformatics support, BRAF


and strict adherence to many-faceted quality control BRAF (B-raf proto oncogene, serine/threonine kinase) is
matrices. Among preanalytical factors, careful sample a signal transduction protein kinase, lying downstream
assessment, suitable quality and quantities of input to KRAS signaling pathway and is mutated across mul-
DNA/RNA, validation of minimal acceptable sequence tiple tumor type including melanoma, lymphoma/leu-
depth coverage, and trained personnel for accurate kemias, and carcinomas. The most widely studied
interpretation are of utmost importance.112 The wide- activating BRAF mutation, viz., Val600Gly (V600E) is
spread implementation of NGS technologies in day- common in melanoma (up to 50%) and colon carci-
to-day clinics remains a work in progress. noma (10%e15%) and is a biomarker of its targetable
inhibitor, vemurafenib.129 BRAF mutations are uncom-
mon in NSCLC and has a reported prevalence of up to
OTHERS DRIVER ONCOGENES 4%. The commonly seen mutations in NSCLC are
MET V600E, and other mutations in exon 11 and 15. The
MET, the gene encoding hepatocyte growth factor, is BRAF-mutated lung tumors, specifically V600E muta-
amplified in 5%e20% of EGFR-TKI-treated relapsed tion, correlated with micropapillary histology and
pulmonary adenocarcinoma. In vitro study hypothe- worse overall survival.130
sized MET amplification may be present as small sub-
clone in primary pulmonary adenocarcinoma.118 RET
While MET amplifications are more common in post- RET rearrangements are rare events in pulmonary
treatment NSCLC,119 the other MET mutation is a skip- adenocarcinoma and has an estimated prevalence of
ping or splice mutation in exon 14. The exon 14 splice w1%e2%.70,131e133 Similar to other RTK fusion tu-
mutation has a reported frequency of w5% and is mors such as ALK and ROS1, RET-rearranged tumors
enriched in wild-type for known driver gene mutation are more common in young patients and never smokers
patients, older age group, female gender, and with to light smokers, and occasionally show poorly differ-
never-smoking history.120e123 Detection for MET alter- entiated adenocarcinoma morphology.134 A number
ations are dependent on sequencing or NGS-based ap- of fusion variants have now been described, with
proaches while MET amplification can be detected by KIF5B-RET being the most common variant.135 RET fu-
FISH.124 Multiple targetable molecules including crizo- sions are typically detected by FISH or by NGS-based
tinib, cabozantinib, and others and multiple clinical tri- methods. Data regarding IHC-based detection are
als are presently underway.124 limited; however, the available antibody clones lack
adequate sensitivity for reliable detection in clinical lab-
KRAS oratories. Recent clinical trial with RET inhibitor cabo-
The most commonly mutated driver oncogene in pul- zantinib has shown preliminary efficacy,136,137
monary adenocarcinoma is KRAS (Kirsten rat sarcoma multiple clinical trials are presently underway.138
2 viral oncogene homolog), a member of the RAS fam-
ily of membrane-associated G-proteins, that exerts its NTRK1
effects on RTKs including EGFR. In contrast to EGFR, NTRK1 (neurotrophic RTK 1) fusions are reported in
KRAS mutations are more common in smokers. In w1%e2% of NSCLC. The first reported NTRK1 fusions
larger series, up to 30%e34% of smokers harbored in a cohort of pulmonary adenocarcinoma were pan-
KRAS mutation49,125 as opposed to w5%e7% in non- negative for all known driver mutations; the identified
smokers. KRAS mutations are more frequent in Cauca- cases were female patients.139 However, larger and inde-
sians than Asians126 and are essentially mutually pendent studies are needed to assess clinicopathologic
exclusive to EGFR mutations or ALK fusions.127 Solid characteristics. The availability of targetable drugs
histology pattern, tumor infiltrating leukocytes, and (such as crizotinib, entrectenib) and early clinical
pulmonary mucinous adenocarcinoma are associated response data with entrectinib are promising.97
with KRAS mutations.20,21 The majority of KRAS muta-
tions occurs in codons 12 and 13.49 The G12A and HER2
G12C mutations are more common in nonmucinous HER2 or ERBB2 (human EGFR 2), located on chromo-
adenocarcinoma while G12V, G12D, G13D mutations some 17 is a member of ERBB family encoded by
are seen in mucinous as well as nonmucinous adeno- erb-b2 RTK and plays key roles in oncogenesis and pro-
carcinoma histology.128 gression of several malignancies. HER2 overexpression
24 Pulmonary Adenocarcinoma: Approaches to Treatment

is reported in breast, stomach, lung, bladder, ovarian DNA compared with healthy individuals.151 The frag-
and pancreatic carcinoma. It is a biomarker and thera- ment lengths of ctDNA are also shorter compared
peutic target in breast and gastric carcinoma and pre- with normal circulating DNA.152 These, among other
dicts response to targetable molecules trastuzumab observations, have led to the exploitation of using
and lapatinib. Majority of the reported HER2 mutations blood samples to detect tumor-specific DNA or ctDNA.
(1%e3% frequency) in NSCLC are small in-frame CtDNA is markedly dilute compared with normal
insertion (exon 20) in its kinase domain, while the circulating DNA and requires highly sensitive methods
transmembrane domain mutation is uncommon of detection. These detection methods can be broadly
(0.18%).140,141 The clinical efficacy of HER2-targeted categorized into two ways: targeting specific molecular
therapy in NSCLC is still evolving.142,143 A recent study alterations and targeting all/multiple possible molecu-
showed promising results with Afatinib, a pan-HER2 in- lar alterations.
hibitor in HER2 transmembrane domain mutation
subgroup.140 Targeting Specific Molecular Alterations
PCR-based detection methods are primarily used to
detect specific molecular alterations, such as specific
LIQUID BIOPSIES EGFR-sensitizing mutations or the T790M resistance
Management of lung adenocarcinomas stands as a hall- mechanism. They obtain specificity by using a
mark example of precision oncology. It is now routine sequence-specific primer, such as ARMS,153,154 or pref-
to test for EGFR-sensitizing mutations and ALK rear- erentially amplifying mismatched sequences to enrich
rangements, with an ever-growing number of action- nontarget sequences of a mixed template, such as
able targets in the pipeline. Consideration of the PNA-clamping.155 A new method called digital PCR,
tissue material for molecular testing is now more crucial greatly improves specificity and sensitivity compared
than ever. Initial diagnostic biopsy material can some- with nondigital methods. This technology involves
times be very limited, and many patients with advanced separating the template mix into tiny individual reac-
disease will not be eligible for surgical treatment and tion vessels, most commonly by forming droplets.
miss the opportunity of acquiring more tissue. An alter- These individual reaction droplets can then be assessed
native to tissue biopsy is the utilization of circulating tu- individually for the presence of the mutation and essen-
mor cells (CTCs) or circulating tumor DNA (ctDNA) to tially converts the analog nature of PCR into a linear
obtain a “liquid biopsy.” digital signal that gives way for absolute quantification
of variant alleles.156
Circulating Tumor Cells
Malignant single cells or aggregates may be found in Targeting Multiple Molecular Alterations
blood once the tumor has invaded vasculature and Increasing affordability of massive parallel sequencing,
entered the circulatory system. These occur at a rate of or NGS, has led to the implementation of cancer-
about 1 in 100 million cells.144 Elevated levels of these specific target gene panels in ctDNA. These panels cover
CTCs have shown significant survival differences in a multitude of genes recurrently mutated in cancer.
multiple cancers,145,146 including small cell lung can- Although still largely used in FFPE tumor tissue,
cer147 and NSCLC.148,149 The United States Food and methods that greatly increase its detection sensitivity
Drug Administration has approved one CTC selection are beginning to allow its use in liquid biopsies.157
platform, the CellSearch System (Janssen Diagnostics,
Raritan, New Jersey). However, overall there remains a Clinical Applications
significant barrier in the clinical implementation of Liquid biopsies may play important roles in patients
CTC in lung cancer due to the generally low number with lung cancer. These can be especially helpful in pa-
of detectable tumor cells. tients with genomically defined tumors, such as those
harboring EGFR-sensitizing mutations. CTCs and
Circulating Tumor DNA ctDNA containing the specific EGFR mutation can
Every living cell secretes small amounts of DNA into the persist in the bloodstream after therapy and during
circulation, and its concentration increases in certain relapse. Currently, liquid biopsies serve three purposes
conditions such as trauma, inflammation, apoptosis, in its clinical applicability. They can be used to (1)
or necrosis.150 Patients with malignant disease have assess tumor burden, (2) characterize resistance mecha-
been found to have a significant increase in circulating nisms, and (3) monitor response to therapy.
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 25

Assessing Tumor Burden Much focus on immune checkpoint inhibition, or


Analogous to measuring HIV viral load, quantification “releasing the brakes,” has been garnered ever since
of circulating DNA can serve as a surrogate for tumor numerous clinical trials have shown remarkably
burden in cancer patients, especially in metastatic increased efficacy of antiimmune checkpoint inhibitor
disease.158e160 Its application in early-stage lung molecules (e.g., anti-CTLA4, anti-PDL1) in treating
adenocarcinoma, however, may not be as noteworthy various cancers, including lung cancer.165,166
as in other histologic subtypes. The recent TRACERx Program death ligand 1 (PD-L1), a member of the
study showed that ctDNA in early-stage lung adenocar- B7 family, is a major immune checkpoint protein that
cinoma was not easily detected compared to non- mediates antitumor immune suppression and response.
adenocarcinoma histology.161 They also showed a In lung tissue, it is expressed on a variety of cells,
correlation between tumor volume and ctDNA variant including macrophages, T-cells, epithelial cells, and
allele frequencies (VAF), where a primary tumor vol- endothelial cells.167 Its ligand, programmed death re-
ume of 10 cm3 predicted a ctDNA plasma VAF of 0.1%. ceptor (PD1), is expressed on T-cells and is upregulated
after activation by a foreign antigen. The interaction of
Characterizing Resistance Mechanisms PD-L1-PD1 serves as an important mediator of the anti-
Patients who progress while on TKI therapy may need inflammatory processes to downregulate inflammatory
another biopsy to characterize the tumor’s resistance signals and prevent collateral tissue damage. Some tu-
mechanism. If the tumor is inaccessible, conventional mors are able to “highjack” this mechanism by express-
methods may possibly carry a significant morbidity ing PD-L1, escaping destruction of activated T-cells.
risk to the patient. Moreover, limited tissue sampling Fortunately, this also serves as a means for intervention
will not always sample all the subclones present to by providing a target for therapy. Many clinical trials
cover the full spectrum of the tumor’s heterogeneity. have established immune checkpoint inhibition, in
Plasma detection of resistance mutations, especially particular via the PD1/PD-L1 axis, as a new therapeutic
T790M, via the significantly less invasive liquid biopsy paradigm in the management of cancer, including lung
is a sensible alternative in this regard.162,163 cancer. PD1/PD-L1 axis agents include PD1 inhibitors
such as nivolumab and pembrolizumab, and PD-L1 in-
Monitoring Response to Therapy hibitors such as atezolizumab, durvalumab, and avelu-
Several groups have demonstrated a role for detecting mab. Discussion for each clinical trial is beyond the
ctDNA EGFR mutations during treatment with an scope of this chapter and details should be sought in
EGFR-TKI.162,164 Because of the quantitative nature of the other chapters.
the different detection methods, in particular digital
PCR, the liquid biopsy is finding utility as a way to Program Death Ligand 1 Testing
monitoring response to therapy. Benefit of anti-PD-L1 immunotherapy is found in only
Although still not ready for routine clinical applica- a subset of patients with lung adenocarcinoma. There-
tion, extensive research for more sensitive and specific fore, testing for tumors for PD-L1 expression via immu-
methods may see liquid biopsy as an essential diag- nohistochemistry serves as an eligibility biomarker used
nostic armamentarium in the clinical management of to select patients who might benefit from anti-PD-L1
lung cancer patients. immunotherapy. It is now a standard clinical practice
for lung adenocarcinomas. Different PD-L1 diagnostic
antibody clones have been developed that correspond
PROGRAM DEATH LIGAND 1 with the different PD1/PD-L1 inhibitors (Table 2.3).
Tumor-associated antigens induce a host immune Histologic evaluation of tumor PD-L1 expression re-
response which, at least in the early stages of disease, quires a few important issues that one needs to be aware
provides a mechanism for control and even destruction of. In lung tissue, PD-L1 expression may be present on
of malignant cells. This important process of immuno- dendritic cells, macrophages, mast cells, T-cells, B-cells,
surveillance is underscored when the immune system is and endothelial cells.168 Examination of the hematoxy-
compromised, such as in HIV patients, or when cancer lin and eosin (H & E)estained slides should be per-
cells evolve acquire various tolerance mechanisms, formed to assess tissue preservation and staining
allowing immune escape. These mechanisms include quality. External positive (human tonsils or cell lines)
inhibition of regulatory T-cells, defective antigen pre- and negative control slides should also be examined.
sentation, immune suppressive mediators, or dysregula- The minimum number for tumor cells needed for ade-
tion of costimulatory and coinhibitory molecules. quacy is set by the manufacturer; 50 cells is needed for
26 Pulmonary Adenocarcinoma: Approaches to Treatment

TABLE 2.3
Immune Checkpoint Inhibitors, Associated PD-L1 Immunohistochemistry Assays, and Characteristics
Checkpoint PD-L1 Antibody Clones
Inhibitor (Host) Epitope Autostainer Detection System
Nivolumab 28-8 (Rabbit monoclonal) Extracellular Dako Link 48 Envision flex
Pembrolizumab 22C3 (Mouse monoclonal) Extracellular
Atezolizumab SP142 (Rabbit monoclonal) Cytoplasmic Ventana Optiview þ amplification
Durvalumab SP263 (Rabbit monoclonal) Cytoplasmic Benchmark Optiview
Avulumab 73-10 (Rabbit monoclonal) Cytoplasmic Dako Envision flex

the SP142 assay (Ventana) and 100 cells is needed for Furthermore, molecular profiling of lung adenocarci-
the 28-8 and 22C3 pharmDx assays (Agilent Technolo- noma has identified multiple genomic aberrations,
gies/Dako). Care needs to be taken not to mistake including mutations and translocations that are drivers
intra-alveolar macrophages for denuded tumor cells. of oncogenicity in these tumors, each of which may be
High-magnification examination is necessary to so as targeted by specific therapy. These have led to increased
not to overlook weak staining (see Figure 2.3). personalization of the treatment of lung adenocarci-
The availability of various PD-L1/PD1 axis drugs, noma patients based on the results of biomarker
each accompanied with a corresponding diagnostic testing for these target aberrations. With rapid advances
antibody clone, not to mention the various cutoffs in our understanding of the complex genetics of lung
established for consideration of PD-L1 positivity, has adenocarcinoma and technologies to assess them, the
made the field quite complex. Definition of positivity classification of lung adenocarcinoma will continue
varies depending on the assay used. When assessing to evolve, with incorporation of novel molecular fea-
for PD-L1 positivity in 28-8, 22C3, SP263, or 73-10 as- tures that drive the biology and treatment of this
says, only partial or complete membranous staining of tumor.
tumor cells should be counted. For the SP142 assay,
both tumor cell staining and immune cell staining are
considered for the criteria of positivity. These differ-
ences make it difficult to standardize one universal REFERENCES
IHC test and/or criteria for establishing PD-L1 positiv- 1. Travis WDBE, Burke AP, Marx A, Nicholson AG. WHO
ity. It is important to note that PD-L1 positivity for Classification of Tumours of Lung, Pleura, Thymus, and Heart.
each assay was established individually for the ability 4th ed. Lyon: IARC; 2015.
2. Travis WD. The 2015 WHO classification of lung tumors.
to predict response to their corresponding drug. Even
Pathologe. 2014;35(suppl 2):188.
so, many studies have been or are currently being un- 3. Travis WD, Brambilla E, Nicholson AG, et al. The 2015
dertaken to unify and establish a technical equivalence World Health Organization Classification of Lung Tu-
between the various assays,169,170 although proving pre- mors: Impact of Genetic, Clinical and Radiologic Ad-
dictive equivalence may be much more difficult. vances Since the 2004 Classification. J Thorac Oncol.
Detailed discussion on PD-L1 immunohistochemistry 2015;10(9):1243e1260.
assays and testing can be found in the IASLC Atlas on 4. Lamb D. Histological classification of lung cancer.
PD-L1 Testing in Lung Cancer.171 Thorax. 1984;39(3):161e165.
5. Shimosato Y, Suzuki A, Hashimoto T, et al. Prognostic
implications of fibrotic focus (scar) in small
CONCLUSIONS peripheral lung cancers. Am J Surg Pathol. 1980;4(4):
365e373.
During the last two decades, adenocarcinoma has
6. Russell PA, Barnett SA, Walkiewicz M, et al. Correlation of
become the most common type of lung cancers. The mutation status and survival with predominant histolog-
2015 World Health Organization classification system ic subtype according to the new IASLC/ATS/ERS lung
for lung adenocarcinoma is not only based on histolog- adenocarcinoma classification in stage III (N2) patients.
ical patterns but also highlights prognostic implication. J Thorac Oncol. 2013;8(4):461e468.
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 27

7. Russell PA, Wainer Z, Wright GM, Daniels M, Conron M, 18. Morales-Oyarvide V, Mino-Kenudson M. High-grade lung
Williams RA. Does lung adenocarcinoma subtype predict adenocarcinomas with micropapillary and/or solid pat-
patient survival?: A clinicopathologic study based on the terns: a review. Curr Opin Pulm Med. 2014;20(4):317e323.
new International Association for the Study of Lung 19. Finberg KE, Sequist LV, Joshi VA, et al. Mucinous differen-
Cancer/American Thoracic Society/European Respiratory tiation correlates with absence of EGFR mutation and
Society international multidisciplinary lung adenocarci- presence of KRAS mutation in lung adenocarcinomas
noma classification. J Thorac Oncol. 2011;6(9): with bronchioloalveolar features. J Mol Diagn. 2007;
1496e1504. 9(3):320e326.
8. Tsuta K, Kawago M, Inoue E, et al. The utility of the pro- 20. Rekhtman N, Ang DC, Riely GJ, Ladanyi M, Moreira AL.
posed IASLC/ATS/ERS lung adenocarcinoma subtypes for KRAS mutations are associated with solid growth
disease prognosis and correlation of driver gene pattern and tumor-infiltrating leukocytes in lung
alterations. Lung Cancer. 2013;81(3):371e376. adenocarcinoma. Modern pathology : an official journal of
9. Travis WD, Brambilla E, Noguchi M, et al. International the United States and Canadian Academy of Pathology, Inc.
association for the study of lung cancer/american 2013;26(10):1307e1319.
thoracic society/european respiratory society interna- 21. Kadota K, Yeh YC, D’Angelo SP, et al. Associations be-
tional multidisciplinary classification of lung tween mutations and histologic patterns of mucin in
adenocarcinoma. J Thorac Oncol. 2011;6(2):244e285. lung adenocarcinoma: invasive mucinous pattern and
10. Yoshizawa A, Motoi N, Riely GJ, et al. Impact of proposed extracellular mucin are associated with KRAS mutation.
IASLC/ATS/ERS classification of lung adenocarcinoma: Am J Surg Pathol. 2014;38(8):1118e1127.
prognostic subgroups and implications for further revi- 22. Hwang DH, Sholl LM, Rojas-Rudilla V, et al. KRAS and
sion of staging based on analysis of 514 stage I cases. NKX2-1 Mutations in Invasive Mucinous Adenocarci-
Modern pathology : an official journal of the United States noma of the Lung. J Thorac Oncol. 2016;11(4):496e503.
and Canadian Academy of Pathology, Inc. 2011;24(5): 23. Duruisseaux M, McLeer-Florin A, Antoine M, et al. NRG1
653e664. fusion in a French cohort of invasive mucinous lung
11. Behera M, Owonikoko TK, Gal AA, et al. Lung Adenocar- adenocarcinoma. Cancer Med. 2016;5(12):3579e3585.
cinoma Staging Using the 2011 IASLC/ATS/ERS Classifi- 24. Fernandez-Cuesta L, Plenker D, Osada H, et al. CD74-
cation: A Pooled Analysis of Adenocarcinoma In Situ NRG1 fusions in lung adenocarcinoma. Cancer Discov.
and Minimally Invasive Adenocarcinoma. Clin Lung Can- 2014;4(4):415e422.
cer. 2016;17(5):e57ee64. 25. Rami-Porta R, Bolejack V, Crowley J, et al. The IASLC
12. Warth A, Muley T, Kossakowski C, et al. Prognostic Lung Cancer Staging Project: Proposals for the Revisions
impact and clinicopathological correlations of the cribri- of the T Descriptors in the Forthcoming Eighth Edition of
form pattern in pulmonary adenocarcinoma. J Thorac the TNM Classification for Lung Cancer. J Thorac Oncol.
Oncol. 2015;10(4):638e644. 2015;10(7):990e1003.
13. Pan Y, Zhang Y, Li Y, et al. ALK, ROS1 and RET fusions in 26. Goldstraw P, Chansky K, Crowley J, et al. The IASLC Lung
1139 lung adenocarcinomas: a comprehensive study of Cancer Staging Project: Proposals for Revision of the
common and fusion pattern-specific clinicopathologic, TNM Stage Groupings in the Forthcoming (Eighth) Edi-
histologic and cytologic features. Lung Cancer. 2014; tion of the TNM Classification for Lung Cancer. J Thorac
84(2):121e126. Oncol. 2016;11(1):39e51.
14. Lee MC, Buitrago DH, Kadota K, Jones DR, 27. Asamura H, Chansky K, Crowley J, et al. The Interna-
Adusumilli PS. Recent advances and clinical implications tional Association for the Study of Lung Cancer Lung
of the micropapillary histological subtype in lung Cancer Staging Project: Proposals for the Revision of
adenocarcinomas. Lung Cancer Manag. 2014;3(3): the N Descriptors in the Forthcoming 8th Edition of
245e253. the TNM Classification for Lung Cancer. J Thorac Oncol.
15. Tsao MS, Marguet S, Le Teuff G, et al. Subtype Classifica- 2015;10(12):1675e1684.
tion of Lung Adenocarcinoma Predicts Benefit From 28. Eberhardt WE, Mitchell A, Crowley J, et al. The IASLC
Adjuvant Chemotherapy in Patients Undergoing Com- Lung Cancer Staging Project: Proposals for the Revision
plete Resection. J Clin Oncol. 2015;33(30):3439e3446. of the M Descriptors in the Forthcoming Eighth Edition
16. Kadota K, Nitadori J, Sima CS, et al. Tumor Spread of the TNM Classification of Lung Cancer. J Thorac Oncol.
through Air Spaces is an Important Pattern of 2015;10(11):1515e1522.
Invasion and Impacts the Frequency and Location of 29. Nicholson AG, Chansky K, Crowley J, et al. The Interna-
Recurrences after Limited Resection for Small Stage I tional Association for the Study of Lung Cancer Lung
Lung Adenocarcinomas. J Thorac Oncol. 2015;10(5): Cancer Staging Project: Proposals for the Revision of
806e814. the Clinical and Pathologic Staging of Small Cell Lung
17. Pyo JS, Kim JH. Clinicopathological Significance of Cancer in the Forthcoming Eighth Edition of the TNM
Micropapillary Pattern in Lung Adenocarcinoma. Pathol Classification for Lung Cancer. J Thorac Oncol. 2016;
Oncol Res. 2018;24(3):547e555. 11(3):300e311.
28 Pulmonary Adenocarcinoma: Approaches to Treatment

30. Travis WD, Asamura H, Bankier AA, et al. The IASLC Lung 41. Yatabe Y, Mitsudomi T, Takahashi T. TTF-1 expression in
Cancer Staging Project: Proposals for Coding T Categories pulmonary adenocarcinomas. Am J Surg Pathol. 2002;
for Subsolid Nodules and Assessment of Tumor Size in 26(6):767e773.
Part-Solid Tumors in the Forthcoming Eighth Edition of 42. Yamada E, Bastie CC. Disruption of Fyn SH3 domain
the TNM Classification of Lung Cancer. J Thorac Oncol. interaction with a proline-rich motif in liver kinase B1 re-
2016;11(8):1204e1223. sults in activation of AMP-activated protein kinase. PLoS
31. Detterbeck FC, Franklin WA, Nicholson AG, et al. The One. 2014;9(2):e89604.
IASLC Lung Cancer Staging Project: Background Data 43. Hemminki A, Tomlinson I, Markie D, et al. Localization
and Proposed Criteria to Distinguish Separate Primary of a susceptibility locus for Peutz-Jeghers syndrome to
Lung Cancers from Metastatic Foci in Patients with Two 19p using comparative genomic hybridization and tar-
Lung Tumors in the Forthcoming Eighth Edition of the geted linkage analysis. Nat Genet. 1997;15(1):87e90.
TNM Classification for Lung Cancer. J Thorac Oncol. 44. Hemminki A, Markie D, Tomlinson I, et al. A serine/thre-
2016;11(5):651e665. onine kinase gene defective in Peutz-Jeghers syndrome.
32. Detterbeck FC, Bolejack V, Arenberg DA, et al. The IASLC Nature. 1998;391(6663):184e187.
Lung Cancer Staging Project: Background Data and Pro- 45. Sanchez-Cespedes M, Parrella P, Esteller M, et al. Inactiva-
posals for the Classification of Lung Cancer with Separate tion of LKB1/STK11 is a common event in adenocarci-
Tumor Nodules in the Forthcoming Eighth Edition of the nomas of the lung. Cancer Res. 2002;62(13):3659e3662.
TNM Classification for Lung Cancer. J Thorac Oncol. 2016; 46. Carretero J, Medina PP, Pio R, Montuenga LM, Sanchez-
11(5):681e692. Cespedes M. Novel and natural knockout lung cancer
33. Detterbeck FC, Marom EM, Arenberg DA, et al. The IASLC cell lines for the LKB1/STK11 tumor suppressor gene.
Lung Cancer Staging Project: Background Data and Pro- Oncogene. 2004;23(22):4037e4040.
posals for the Application of TNM Staging Rules to 47. Gill RK, Yang SH, Meerzaman D, et al. Frequent homozy-
Lung Cancer Presenting as Multiple Nodules with gous deletion of the LKB1/STK11 gene in non-small cell
Ground Glass or Lepidic Features or a Pneumonic Type lung cancer. Oncogene. 2011;30(35):3784e3791.
of Involvement in the Forthcoming Eighth Edition of 48. Dumstrei K, Nassif C, Abboud G, Aryai A, Aryai A,
the TNM Classification. J Thorac Oncol. 2016;11(5): Hartenstein V. EGFR signaling is required for the differen-
666e680. tiation and maintenance of neural progenitors along the
34. Detterbeck FC, Nicholson AG, Franklin WA, et al. The dorsal midline of the Drosophila embryonic head.
IASLC Lung Cancer Staging Project: Summary of Pro- Development. 1998;125(17):3417e3426.
posals for Revisions of the Classification of Lung Cancers 49. Dogan S, Shen R, Ang DC, et al. Molecular epidemiology of
with Multiple Pulmonary Sites of Involvement in the EGFR and KRAS mutations in 3,026 lung adenocarcinomas:
Forthcoming Eighth Edition of the TNM Classification. higher susceptibility of women to smoking-related KRAS-
J Thorac Oncol. 2016;11(5):639e650. mutant cancers. Clin Cancer Res. 2012;18(22):6169e6177.
35. Cancer Genome Atlas Research N. Comprehensive mo- 50. Shi Y, Au JS, Thongprasert S, et al. A prospective, molec-
lecular profiling of lung adenocarcinoma. Nature. 2014; ular epidemiology study of EGFR mutations in Asian pa-
511(7511):543e550. tients with advanced non-small-cell lung cancer of
36. Bhattacharjee A, Richards WG, Staunton J, et al. Classifi- adenocarcinoma histology (PIONEER). J Thorac Oncol.
cation of human lung carcinomas by mRNA expression 2014;9(2):154e162.
profiling reveals distinct adenocarcinoma subclasses. 51. Yun CH, Mengwasser KE, Toms AV, et al. The T790M mu-
Proc Natl Acad Sci U S A. 2001;98(24):13790e13795. tation in EGFR kinase causes drug resistance by increasing
37. Beer DG, Kardia SL, Huang CC, et al. Gene-expression the affinity for ATP. Proc Natl Acad Sci U S A. 2008;
profiles predict survival of patients with lung 105(6):2070e2075.
adenocarcinoma. Nat Med. 2002;8(8):816e824. 52. Pao W, Miller VA, Politi KA, et al. Acquired resistance of
38. Hayes DN, Monti S, Parmigiani G, et al. Gene expression lung adenocarcinomas to gefitinib or erlotinib is associ-
profiling reveals reproducible human lung adenocarci- ated with a second mutation in the EGFR kinase
noma subtypes in multiple independent patient domain. PLoS Med. 2005;2(3):e73.
cohorts. J Clin Oncol. 2006;24(31):5079e5090. 53. Wu SG, Liu YN, Tsai MF, et al. The mechanism of ac-
39. Wilkerson MD, Yin X, Walter V, et al. Differential quired resistance to irreversible EGFR tyrosine kinase in-
pathogenesis of lung adenocarcinoma subtypes hibitor-afatinib in lung adenocarcinoma patients.
involving sequence mutations, copy number, chromo- Oncotarget. 2016;7(11):12404e12413.
somal instability, and methylation. PLoS One. 2012; 54. Balak MN, Gong Y, Riely GJ, et al. Novel D761Y and com-
7(5):e36530. mon secondary T790M mutations in epidermal growth
40. Garber ME, Troyanskaya OG, Schluens K, et al. Diversity factor receptor-mutant lung adenocarcinomas with ac-
of gene expression in adenocarcinoma of the lung. Proc quired resistance to kinase inhibitors. Clin Cancer Res.
Natl Acad Sci U S A. 2001;98(24):13784e13789. 2006;12(21):6494e6501.
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 29

55. Costa DB, Schumer ST, Tenen DG, Kobayashi S. Differen- 70. Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and
tial responses to erlotinib in epidermal growth factor re- ALK fusions in lung cancer. Nat Med. 2012;18(3):
ceptor (EGFR)-mutated lung cancers with acquired 378e381.
resistance to gefitinib carrying the L747S or T790M 71. Takeuchi K, Choi YL, Togashi Y, et al. KIF5B-ALK, a novel
secondary mutations. J Clin Oncol. 2008;26(7): fusion oncokinase identified by an immunohistochem-
1182e1184; author reply 1184-1186. istry-based diagnostic system for ALK-positive lung
56. Ercan D, Zejnullahu K, Yonesaka K, et al. Amplification of cancer. Clin Cancer Res. 2009;15(9):3143e3149.
EGFR T790M causes resistance to an irreversible EGFR 72. Tsao MS, Hirsch F, Yatabe Y. IASLC Atlas of ALK and ROS1
inhibitor. Oncogene. 2010;29(16):2346e2356. Testing in Lung Cancer. 2nd ed. Editorial Rx Press; 2016.
57. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET 73. Shaw AT, Yeap BY, Solomon BJ, et al. Effect of crizotinib
amplification leads to gefitinib resistance in lung cancer on overall survival in patients with advanced non-small-
by activating ERBB3 signaling. Science. 2007;316(5827): cell lung cancer harbouring ALK gene rearrangement: a
1039e1043. retrospective analysis. Lancet Oncol. 2011;12(11):
58. Ohashi K, Maruvka YE, Michor F, Pao W. Epidermal 1004e1012.
growth factor receptor tyrosine kinase inhibitor-resistant 74. Shaw AT, Yeap BY, Mino-Kenudson M, et al. Clinical fea-
disease. J Clin Oncol. 2013;31(8):1070e1080. tures and outcome of patients with non-small-cell lung
59. Camidge DR, Pao W, Sequist LV. Acquired resistance to cancer who harbor EML4-ALK. J Clin Oncol. 2009;
TKIs in solid tumours: learning from lung cancer. Nat 27(26):4247e4253.
Rev Clin Oncol. 2014;11(8):473e481. 75. Rodig SJ, Mino-Kenudson M, Dacic S, et al. Unique clin-
60. Rosell R, Karachaliou N, Morales-Espinosa D, et al. Adap- icopathologic features characterize ALK-rearranged lung
tive resistance to targeted therapies in cancer. Transl Lung adenocarcinoma in the western population. Clin Cancer
Cancer Res. 2013;2(3):152e159. Res. 2009;15(16):5216e5223.
61. Yu HA, Riely GJ, Lovly CM. Therapeutic strategies utilized in 76. Lindeman NI, Cagle PT, Beasley MB, et al. Molecular
the setting of acquired resistance to EGFR tyrosine kinase testing guideline for selection of lung cancer patients
inhibitors. Clin Cancer Res. 2014;20(23):5898e5907. for EGFR and ALK tyrosine kinase inhibitors: guideline
62. Oser MG, Niederst MJ, Sequist LV, Engelman JA. Trans- from the College of American Pathologists, International
formation from non-small-cell lung cancer to small-cell Association for the Study of Lung Cancer, and Associa-
lung cancer: molecular drivers and cells of origin. The tion for Molecular Pathology. J Thorac Oncol. 2013;8(7):
Lancet Oncology. 2015;16(4):e165ee172. 823e859.
63. Niederst MJ, Sequist LV, Poirier JT, et al. RB loss in resis- 77. Alrifai D, Popat S, Ahmed M, et al. A rare case of squa-
tant EGFR mutant lung adenocarcinomas that transform mous cell carcinoma of the lung harbouring ALK and
to small-cell lung cancer. Nat Commun. 2015;6:6377. BRAF activating mutations. Lung Cancer. 2013;80(3):
64. Stewart EL, Tan SZ, Liu G, Tsao MS. Known and putative 339e340.
mechanisms of resistance to EGFR targeted therapies in 78. Ochi N, Yamane H, Yamagishi T, Takigawa N, Monobe Y.
NSCLC patients with EGFR mutations-a review. Transl Can we eliminate squamous cell carcinoma of the lung
Lung Cancer Res. 2015;4(1):67e81. from testing of EML4-ALK fusion gene? Lung Cancer.
65. Morris SW, Kirstein MN, Valentine MB, et al. Fusion of a 2013;79(1):94e95.
kinase gene, ALK, to a nucleolar protein gene, NPM, in 79. Lovly CM, Pao W. Escaping ALK inhibition: mechanisms
non-Hodgkin’s lymphoma. Science. 1994;263(5151): of and strategies to overcome resistance. Sci Transl Med.
1281e1284. 2012;4(120):120e122.
66. Soda M, Choi YL, Enomoto M, et al. Identification of the 80. Lovly CM, Shaw AT. Molecular pathways: resistance to ki-
transforming EML4-ALK fusion gene in non-small-cell nase inhibitors and implications for therapeutic
lung cancer. Nature. 2007;448(7153):561e566. strategies. Clin Cancer Res. 2014;20(9):2249e2256.
67. Rikova K, Guo A, Zeng Q, et al. Global survey of phos- 81. van der Wekken AJ, Saber A, Hiltermann TJ, Kok K, van
photyrosine signaling identifies oncogenic kinases in den Berg A, Groen HJ. Resistance mechanisms after tyro-
lung cancer. Cell. 2007;131(6):1190e1203. sine kinase inhibitors afatinib and crizotinib in non-
68. Ou SH, Klempner SJ, Greenbowe JR, et al. Identification small cell lung cancer, a review of the literature. Crit Rev
of a novel HIP1-ALK fusion variant in Non-Small-Cell Oncol Hematol. 2016;100:107e116.
Lung Cancer (NSCLC) and discovery of ALK I1171 82. Sasaki T, Koivunen J, Ogino A, et al. A novel ALK
(I1171N/S) mutations in two ALK-rearranged NSCLC pa- secondary mutation and EGFR signaling cause resistance
tients with resistance to Alectinib. J Thorac Oncol. 2014; to ALK kinase inhibitors. Cancer Res. 2011;71(18):
9(12):1821e1825. 6051e6060.
69. Hong S, Fang W, Hu Z, et al. A large-scale cross-sectional 83. Robinson DR, Wu YM, Lin SF. The protein tyrosine kinase
study of ALK rearrangements and EGFR mutations in family of the human genome. Oncogene. 2000;19(49):
non-small-cell lung cancer in Chinese Han population. 5548e5557.
Sci Rep. 2014;4:7268.
30 Pulmonary Adenocarcinoma: Approaches to Treatment

84. Birchmeier C, Sharma S, Wigler M. Expression and From Members of the Pulmonary Pathology Society. Ar-
rearrangement of the ROS1 gene in human glioblastoma chives of pathology & laboratory medicine. 2018;142(3):
cells. Proc Natl Acad Sci U S A. 1987;84(24):9270e9274. 408e419.
85. Charest A, Lane K, McMahon K, et al. Fusion of FIG to the 100. Bubendorf L, Buttner R, Al-Dayel F, et al. Testing for
receptor tyrosine kinase ROS in a glioblastoma with an ROS1 in non-small cell lung cancer: a review with
interstitial del(6)(q21q21). Genes Chromosomes Cancer. recommendations. Virchows Arch. 2016;469(5):
2003;37(1):58e71. 489e503.
86. Bergethon K, Shaw AT, Ou SH, et al. ROS1 rearrange- 101. Lee SE, Lee B, Hong M, et al. Comprehensive analysis of
ments define a unique molecular class of lung cancers. RET and ROS1 rearrangement in lung adenocarcinoma.
J Clin Oncol. 2012;30(8):863e870. Modern pathology : an official journal of the United States
87. Pal P, Khan Z. Ros1-1. J Clin Pathol. 2017;70(12): and Canadian Academy of Pathology, Inc. 2015;28(4):
1001e1009. 468e479.
88. Zhu Q, Zhan P, Zhang X, Lv T, Song Y. Clinicopathologic 102. Shan L, Lian F, Guo L, et al. Detection of ROS1 gene rear-
characteristics of patients with ROS1 fusion gene in non- rangement in lung adenocarcinoma: comparison of IHC,
small cell lung cancer: a meta-analysis. Transl Lung Cancer FISH and real-time RT-PCR. PLoS One. 2015;10(3):
Res. 2015;4(3):300e309. e0120422.
89. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1- 103. Cao B, Wei P, Liu Z, et al. Detection of lung adenocarci-
rearranged non-small-cell lung cancer. N Engl J Med. noma with ROS1 rearrangement by IHC, FISH, and RT-
2014;371(21):1963e1971. PCR and analysis of its clinicopathologic features. Onco
90. Mazieres J, Zalcman G, Crino L, et al. Crizotinib therapy Targets Ther. 2016;9:131e138.
for advanced lung adenocarcinoma and a ROS1 rear- 104. Suehara Y, Arcila M, Wang L, et al. Identification of
rangement: results from the EUROS1 cohort. J Clin Oncol. KIF5B-RET and GOPC-ROS1 fusions in lung adenocarci-
2015;33(9):992e999. nomas through a comprehensive mRNA-based screen for
91. Zhao J, Zheng J, Kong M, Zhou J, Ding W, Zhou J. tyrosine kinase fusions. Clin Cancer Res. 2012;18(24):
Advanced lung adenocarcinomas with ROS1-rearrange- 6599e6608.
ment frequently show hepatoid cell. Oncotarget. 2016; 105. Dama E, Tillhon M, Bertalot G, et al. Sensitive and afford-
7(45):74162e74170. able diagnostic assay for the quantitative detection of
92. Yoshida A, Tsuta K, Wakai S, et al. Immunohistochemical anaplastic lymphoma kinase (ALK) alterations in pa-
detection of ROS1 is useful for identifying ROS1 rear- tients with non-small cell lung cancer. Oncotarget. 2016;
rangements in lung cancers. Modern pathology : an official 7(24):37160e37176.
journal of the United States and Canadian Academy of Pathol- 106. Lira ME, Choi YL, Lim SM, et al. A single-tube multi-
ogy, Inc. 2014;27(5):711e720. plexed assay for detecting ALK, ROS1, and RET fusions
93. Shaw AT, Hsu PP, Awad MM, Engelman JA. Tyrosine ki- in lung cancer. J Mol Diagn. 2014;16(2):229e243.
nase gene rearrangements in epithelial malignancies. 107. Mino-Kenudson M, Chirieac LR, Law K, et al. A novel,
Nat Rev Cancer. 2013;13(11):772e787. highly sensitive antibody allows for the routine detection
94. Awad MM, Katayama R, McTigue M, et al. Acquired resis- of ALK-rearranged lung adenocarcinomas by standard
tance to crizotinib from a mutation in CD74-ROS1. immunohistochemistry. Clin Cancer Res. 2010;16(5):
N Engl J Med. 2013;368(25):2395e2401. 1561e1571.
95. Facchinetti F, Loriot Y, Kuo MS, et al. Crizotinib-Resistant 108. Cutz JC, Craddock KJ, Torlakovic E, et al. Canadian
ROS1 Mutations Reveal a Predictive Kinase Inhibitor anaplastic lymphoma kinase study: a model for multi-
Sensitivity Model for ROS1- and ALK-Rearranged Lung center standardization and optimization of ALK testing
Cancers. Clin Cancer Res. 2016;22(24):5983e5991. in lung cancer. J Thorac Oncol. 2014;9(9):1255e1263.
96. Dagogo-Jack I, Shaw AT. Expanding the Roster of ROS1 109. Rimkunas VM, Crosby KE, Li D, et al. Analysis of receptor
Inhibitors. J Clin Oncol. 2017;35(23):2595e2597. tyrosine kinase ROS1-positive tumors in non-small cell
97. Drilon A, Siena S, Ou SI, et al. Safety and Antitumor Ac- lung cancer: identification of a FIG-ROS1 fusion. Clin
tivity of the Multitargeted Pan-TRK, ROS1, and ALK In- Cancer Res. 2012;18(16):4449e4457.
hibitor Entrectinib: Combined Results from Two Phase 110. Sholl LM, Sun H, Butaney M, et al. ROS1 immunohisto-
I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. chemistry for detection of ROS1-rearranged lung
2017;7(4):400e409. adenocarcinomas. Am J Surg Pathol. 2013;37(9):1441e1449.
98. Chong CR, Bahcall M, Capelletti M, et al. Identification of 111. Suh JH, Johnson A, Albacker L, et al. Comprehensive
Existing Drugs That Effectively Target NTRK1 and ROS1 Genomic Profiling Facilitates Implementation of the Na-
Rearrangements in Lung Cancer. Clin Cancer Res. 2017; tional Comprehensive Cancer Network Guidelines for
23(1):204e213. Lung Cancer Biomarker Testing and Identifies Patients
99. Thunnissen E, Allen TC, Adam J, et al. Immunohisto- Who May Benefit From Enrollment in Mechanism-
chemistry of Pulmonary Biomarkers: A Perspective Driven Clinical Trials. Oncologist. 2016;21(6):684e691.
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 31

112. Goswami RS, Luthra R, Singh RR, et al. Identification of 125. Slebos RJ, Kibbelaar RE, Dalesio O, et al. K-ras oncogene
Factors Affecting the Success of Next-Generation activation as a prognostic marker in adenocarcinoma of
Sequencing Testing in Solid Tumors. Am J Clin Pathol. the lung. N Engl J Med. 1990;323(9):561e565.
2016;145(2):222e237. 126. Riely GJ, Kris MG, Rosenbaum D, et al. Frequency and
113. Hovelson DH, McDaniel AS, Cani AK, et al. Development distinctive spectrum of KRAS mutations in never smokers
and validation of a scalable next-generation sequencing with lung adenocarcinoma. Clin Cancer Res. 2008;14(18):
system for assessing relevant somatic variants in solid 5731e5734.
tumors. Neoplasia. 2015;17(4):385e399. 127. Gainor JF, Varghese AM, Ou SH, et al. ALK rearrange-
114. Drilon A, Wang L, Arcila ME, et al. Broad, Hybrid Cap- ments are mutually exclusive with mutations in EGFR
ture-Based Next-Generation Sequencing Identifies or KRAS: an analysis of 1,683 patients with non-small
Actionable Genomic Alterations in Lung Adenocarci- cell lung cancer. Clin Cancer Res. 2013;19(15):
nomas Otherwise Negative for Such Alterations by Other 4273e4281.
Genomic Testing Approaches. Clin Cancer Res. 2015; 128. Lee B, Lee T, Lee SH, Choi YL, Han J. Clinicopathologic
21(16):3631e3639. characteristics of EGFR, KRAS, and ALK alterations in
115. Zheng Z, Liebers M, Zhelyazkova B, et al. Anchored 6,595 lung cancers. Oncotarget. 2016;7(17):23874e23884.
multiplex PCR for targeted next-generation sequencing. 129. Salama AK, Flaherty KT. BRAF in melanoma: current stra-
Nat Med. 2014;20(12):1479e1484. tegies and future directions. Clin Cancer Res. 2013;
116. Ali SM, Hensing T, Schrock AB, et al. Comprehensive 19(16):4326e4334.
Genomic Profiling Identifies a Subset of Crizotinib- 130. Marchetti A, Felicioni L, Malatesta S, et al. Clinical fea-
Responsive ALK-Rearranged Non-Small Cell Lung Cancer tures and outcome of patients with non-small-cell lung
Not Detected by Fluorescence In Situ Hybridization. cancer harboring BRAF mutations. J Clin Oncol. 2011;
Oncologist. 2016;21(6):762e770. 29(26):3574e3579.
117. Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan 131. Michels S, Scheel AH, Scheffler M, et al. Clinicopatholog-
Kettering-Integrated Mutation Profiling of Actionable ical Characteristics of RET Rearranged Lung Cancer in Eu-
Cancer Targets (MSK-IMPACT): A Hybridization Cap- ropean Patients. J Thorac Oncol. 2016;11(1):122e127.
ture-Based Next-Generation Sequencing Clinical Assay 132. Wang R, Hu H, Pan Y, et al. RET fusions define a unique
for Solid Tumor Molecular Oncology. J Mol Diagn. molecular and clinicopathologic subtype of non-small-
2015;17(3):251e264. cell lung cancer. J Clin Oncol. 2012;30(35):4352e4359.
118. Turke AB, Zejnullahu K, Wu YL, et al. Preexistence and 133. Tsuta K, Kohno T, Yoshida A, et al. RET-rearranged non-
clonal selection of MET amplification in EGFR mutant small-cell lung carcinoma: a clinicopathological and mo-
NSCLC. Cancer cell. 2010;17(1):77e88. lecular analysis. Br J Cancer. 2014;110(6):1571e1578.
119. Bean J, Brennan C, Shih JY, et al. MET amplification oc- 134. Tsai TH, Wu SG, Hsieh MS, Yu CJ, Yang JC, Shih JY. Clin-
curs with or without T790M mutations in EGFR mutant ical and prognostic implications of RET rearrangements
lung tumors with acquired resistance to gefitinib or in metastatic lung adenocarcinoma patients with malig-
erlotinib. Proc Natl Acad Sci U S A. 2007;104(52): nant pleural effusion. Lung Cancer. 2015;88(2):208e214.
20932e20937. 135. Dugay F, Llamas-Gutierrez F, Gournay M, et al. Clinico-
120. Zheng D, Wang R, Ye T, et al. MET exon 14 skipping de- pathological characteristics of ROS1- and RET-rearranged
fines a unique molecular class of non-small cell lung NSCLC in caucasian patients: Data from a cohort of 713
cancer. Oncotarget. 2016;7(27):41691e41702. non-squamous NSCLC lacking KRAS/EGFR/HER2/BRAF/
121. Heist RS, Shim HS, Gingipally S, et al. MET Exon 14 Skip- PIK3CA/ALK alterations. Oncotarget. 2017;8(32):
ping in Non-Small Cell Lung Cancer. Oncologist. 2016; 53336e53351.
21(4):481e486. 136. Drilon A, Rekhtman N, Arcila M, et al. Cabozantinib in
122. Paik PK, Drilon A, Fan PD, et al. Response to MET inhib- patients with advanced RET-rearranged non-small-cell
itors in patients with stage IV lung adenocarcinomas lung cancer: an open-label, single-centre, phase 2, sin-
harboring MET mutations causing exon 14 skipping. gle-arm trial. Lancet Oncol. 2016;17(12):1653e1660.
Cancer Discov. 2015;5(8):842e849. 137. Drilon A, Wang L, Hasanovic A, et al. Response to Cabo-
123. Awad MM, Oxnard GR, Jackman DM, et al. MET Exon 14 zantinib in patients with RET fusion-positive lung
Mutations in Non-Small-Cell Lung Cancer Are Associated adenocarcinomas. Cancer Discov. 2013;3(6):630e635.
With Advanced Age and Stage-Dependent MET Genomic 138. Song M, Kim SH, Yoon SK. Cabozantinib for the treat-
Amplification and c-Met Overexpression. J Clin Oncol. ment of non-small cell lung cancer with KIF5B-RET
2016;34(7):721e730. fusion. An example of swift repositioning. Arch Pharm
124. Reungwetwattana T, Liang Y, Zhu V, Ou SI. The race to Res. 2015;38(12):2120e2123.
target MET exon 14 skipping alterations in non-small 139. Vaishnavi A, Capelletti M, Le AT, et al. Oncogenic and
cell lung cancer: The Why, the How, the Who, the Un- drug-sensitive NTRK1 rearrangements in lung cancer.
known, and the Inevitable. Lung Cancer. 2017;103: Nat Med. 2013;19(11):1469e1472.
27e37.
32 Pulmonary Adenocarcinoma: Approaches to Treatment

140. Ou SI, Schrock AB, Bocharov EV, et al. HER2 Transmem- non-small-cell lung cancer. Clin Cancer Res. 2006;
brane Domain (TMD) Mutations (V659/G660) That Sta- 12(13):3915e3921.
bilize Homo- and Heterodimerization Are Rare 155. Won JK, Keam B, Koh J, et al. Concomitant ALK translo-
Oncogenic Drivers in Lung Adenocarcinoma That cation and EGFR mutation in lung cancer: a comparison
Respond to Afatinib. J Thorac Oncol. 2017;12(3):446e457. of direct sequencing and sensitive assays and the impact
141. Ettinger DS, Wood DE, Akerley W, et al. Non-Small Cell on responsiveness to tyrosine kinase inhibitor. Ann
Lung Cancer, Version 6.2015. J Natl Compr Canc Netw. Oncol. 2015;26(2):348e354.
2015;13(5):515e524. 156. Vogelstein B, Kinzler KW. Digital PCR. Proc Natl Acad Sci
142. De Greve J, Moran T, Graas MP, et al. Phase II study of U S A. 1999;96(16):9236e9241.
afatinib, an irreversible ErbB family blocker, in demo- 157. Newman AM, Lovejoy AF, Klass DM, et al. Integrated dig-
graphically and genotypically defined lung ital error suppression for improved detection of circu-
adenocarcinoma. Lung Cancer. 2015;88(1):63e69. lating tumor DNA. Nat Biotechnol. 2016;34(5):547e555.
143. De Greve J, Teugels E, Geers C, et al. Clinical activity of 158. Gautschi O, Bigosch C, Huegli B, et al. Circulating deox-
afatinib (BIBW 2992) in patients with lung adenocarci- yribonucleic Acid as prognostic marker in non-small-cell
noma with mutations in the kinase domain of HER2/ lung cancer patients undergoing chemotherapy. J Clin
neu. Lung Cancer. 2012;76(1):123e127. Oncol. 2004;22(20):4157e4164.
144. Kim MY, Oskarsson T, Acharyya S, et al. Tumor self-seed- 159. Tissot C, Toffart AC, Villar S, et al. Circulating free DNA
ing by circulating cancer cells. Cell. 2009;139(7): concentration is an independent prognostic biomarker
1315e1326. in lung cancer. Eur Respir J. 2015;46(6):1773e1780.
145. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tu- 160. Wei Z, Shah N, Deng C, Xiao X, Zhong T, Li X. Circulating
mor cells, disease progression, and survival in metastatic DNA addresses cancer monitoring in non small cell lung
breast cancer. N Engl J Med. 2004;351(8):781e791. cancer patients for detection and capturing the dynamic
146. Cohen SJ, Punt CJ, Iannotti N, et al. Relationship of circu- changes of the disease. Springerplus. 2016;5:531.
lating tumor cells to tumor response, progression-free 161. Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic
survival, and overall survival in patients with metastatic ctDNA analysis depicts early stage lung cancer
colorectal cancer. J Clin Oncol. 2008;26(19):3213e3221. evolution. Nature. 2017;545(7655):446e451.
147. Hou JM, Krebs MG, Lancashire L, et al. Clinical signifi- 162. Mok T, Wu YL, Lee JS, et al. Detection and Dynamic
cance and molecular characteristics of circulating tumor Changes of EGFR Mutations from Circulating Tumor
cells and circulating tumor microemboli in patients DNA as a Predictor of Survival Outcomes in NSCLC Pa-
with small-cell lung cancer. J Clin Oncol. 2012;30(5): tients Treated with First-line Intercalated Erlotinib and
525e532. Chemotherapy. Clin Cancer Res. 2015;21(14):
148. Zhou J, Dong F, Cui F, Xu R, Tang X. The role of circu- 3196e3203.
lating tumor cells in evaluation of prognosis and treat- 163. Thress KS, Brant R, Carr TH, et al. EGFR mutation detec-
ment response in advanced non-small-cell lung cancer. tion in ctDNA from NSCLC patient plasma: A cross-
Cancer Chemother Pharmacol. 2017;79(4):825e833. platform comparison of leading technologies to support
149. Krebs MG, Sloane R, Priest L, et al. Evaluation and prog- the clinical development of AZD9291. Lung Cancer.
nostic significance of circulating tumor cells in patients 2015;90(3):509e515.
with non-small-cell lung cancer. J Clin Oncol. 2011; 164. Lee JY, Qing X, Xiumin W, et al. Longitudinal monitoring
29(12):1556e1563. of EGFR mutations in plasma predicts outcomes of
150. Butt AN, Swaminathan R. Overview of circulating nucleic NSCLC patients treated with EGFR TKIs: Korean Lung
acids in plasma/serum. Ann N Y Acad Sci. 2008;1137: Cancer Consortium (KLCC-12-02). Oncotarget. 2016;
236e242. 7(6):6984e6993.
151. Zaher ER, Anwar MM, Kohail HM, El-Zoghby SM, Abo- 165. Pardoll DM. The blockade of immune checkpoints in
El-Eneen MS. Cell-free DNA concentration and integrity cancer immunotherapy. Nat Rev Cancer. 2012;12(4):
as a screening tool for cancer. Indian J Cancer. 2013; 252e264.
50(3):175e183. 166. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1
152. Underhill HR, Kitzman JO, Hellwig S, et al. Fragment blockade: new immunotherapeutic modalities with dura-
Length of Circulating Tumor DNA. PLoS Genet. 2016; ble clinical benefit in melanoma patients. Clin Cancer Res.
12(7):e1006162. 2013;19(19):5300e5309.
153. Whitcombe D, Theaker J, Guy SP, Brown T, Little S. 167. Yu H, Boyle TA, Zhou C, Rimm DL, Hirsch FR. PD-L1
Detection of PCR products using self-probing amplicons Expression in Lung Cancer. J Thorac Oncol. 2016;11(7):
and fluorescence. Nat Biotechnol. 1999;17(8):804e807. 964e975.
154. Kimura H, Kasahara K, Kawaishi M, et al. Detection of 168. Ai B, Liu H, Huang Y, Peng P. Circulating cell-free DNA as
epidermal growth factor receptor mutations in serum as a prognostic and predictive biomarker in non-small cell
a predictor of the response to gefitinib in patients with lung cancer. Oncotarget. 2016;7(28):44583e44595.
CHAPTER 2 Pulmonary AdenocarcinomadPathology and Molecular Testing 33

169. Hirsch FR, McElhinny A, Stanforth D, et al. PD-L1 Immu- Ligand 1 (PD-L1) on Non-Small Cell Lung Carcinoma.
nohistochemistry Assays for Lung Cancer: Results from Appl Immunohistochem Mol Morphol. 2018;26(2):83e93.
Phase 1 of the Blueprint PD-L1 IHC Assay Comparison 171. Tsao MS, Kerr KM, Dacic S, Yatabe Y, Hirsch F. IASLC
Project. J Thorac Oncol. 2017;12(2):208e222. Atlas of PD-L1 Immunohistochemistry Testing in Lung
170. Parra ER, Villalobos P, Mino B, Rodriguez-Canales J. Cancer. Editorial Rx Press; 2017.
Comparison of Different Antibody Clones for Immuno-
histochemistry Detection of Programmed Cell Death
CHAPTER 3

Adjuvant and Neoadjuvant Therapy in


NoneSmall-Cell Lung Cancer
JEFFREY R. ZWEIG, MD • HEATHER A. WAKELEE, MD

INTRODUCTION regard to survival benefit, with small sample size and se-
The use of adjuvant therapy in patients with none lection bias playing a large role. A 1991 national
small-cell lung cancer (NSCLC) applies to patients consensus report drew the conclusion that based on
with surgically resected stage IB, II, and IIIA disease as available data, there was an unproven benefit of postop-
based on the Tumor, Node, Metastasis staging system. erative chemotherapy and any use of it was considered
While the majority of those with stage IB and II NSCLC experimental.4 In 1995 the Non-small Cell Lung Cancer
are cured, 30%e40% still relapse. The use of adjuvant Collaborative Group published a large metaanalysis on
platinum-based chemotherapy in these patient groups 9387 patients from 52 randomized clinical trials.5 In
results in an absolute survival benefit at 5 years of the 14 trials comparing surgery alone to surgery plus
approximately 5%, with the greatest benefit seen in pa- adjuvant chemotherapy, they found that cisplatin-
tients with stage II and IIIA disease.1 This is the only based chemotherapy regimens (cisplatin, doxorubicin,
treatment that has been proven to improve cure rates af- cyclophosphamide [CAP], or cisplatin plus vindesine)
ter resection, albeit with a modest benefit. Complete as compared with older long-term alkylating agents
surgical resection remains the standard of care in (mainly CAP and nitrosourea) in combination with sur-
early-stage NSCLC.2 With more advanced stage disease, gery, showed a nonstatistically significant absolute sur-
5-year survival rates decrease significantly, with around vival benefit of 5% at 5 years (hazard ratio [HR] 0.87,
66% of patients with stage IB disease alive at 5 years 95% confidence interval [CI] 0.74e1.02, P ¼ .08).
versus 36% of patients with stage IIIA disease.3 There- Long-term alkylating regimens, however, actually
fore there remains an important need to improve on showed chemotherapy in combination with surgery to
these outcomes and identify better postsurgical thera- be detrimental compared with surgery alone, with a
pies for individual patients. 15% increase in the risk of death.5 Although not statis-
Most research to date in the adjuvant setting has tically significant, this trend toward improved overall
focused on the use of chemotherapy. However, interest survival (OS) with a cisplatin-based chemotherapy
in the last years has grown significantly in the use of regimen after surgery was encouraging and paved the
molecularly targeted agents and immunotherapies in way for larger randomized phase III trials to further
early-stage disease, given the significant benefit seen investigate the role of adjuvant cisplatin-based
with these drugs in the unresectable and/or metastatic treatment.
setting. Herein, the current landscape of adjuvant and The results of the first several randomized phase III
neoadjuvant therapy will be discussed, with attention to trials trying to validate these findings with adjuvant
established data regarding the use of chemotherapy and platinum-based regimens were negative. The Eastern
radiation, along with the evolving data regarding the Cooperative Oncology Group (ECOG) 3590 study
use of targeted agents, immunotherapy, and improved di- (Intergroup 0115) was a randomized phase III trial of
agnostics such as circulating tumor DNA (ctDNA). 488 patients after resection of stage II or IIIA NSCLC,
assessing the use of adjuvant radiation versus concur-
rent chemoradiation with cisplatin and etoposide for
ADJUVANT CHEMOTHERAPY 4 planned cycles.6 The primary endpoint was OS. The
Up until 1995, most clinical trials of adjuvant chemo- results were published in 2000 and failed to show a dif-
therapy in operable NSCLC were inconclusive with ference between these two groups. The median OS was

Pulmonary Adenocarcinoma: Approaches to Treatment. https://doi.org/10.1016/B978-0-323-55433-6.00003-1


Copyright © 2019 Elsevier Inc. All rights reserved. 35
36 Pulmonary Adenocarcinoma: Approaches to Treatment

39 months (95% CI 30e52) in the radiation alone published in 2004.9 Here 1867 patients with resected
group versus 38 months (95% CI 31e42) in the chemo- stage I, II, and III NSCLC were randomized to either 3
radiation group (P ¼ .56). There was also no difference or 4 cycles of cisplatin-based chemotherapy or observa-
in treatment effect when subgroups were compared ac- tion. At randomization, 36.5% had stage I disease,
cording to age (younger than 60 years or older than 24.2% had stage II disease, and 39.3% had stage III dis-
60 years), sex, race, stage (II vs. IIIA), ECOG perfor- ease. Chemotherapy regimens included cisplatin plus
mance status (PS; 0 vs. 1), and the number of opera- either etoposide, vinorelbine, vinblastine or vindesine,
tions performed (1 vs. greater than 2). Notably, 31% with etoposide and vinorelbine given to the majority
of patients in the combined chemotherapy plus radia- of patients (56.5% and 26.8%, respectively). A quarter
tion group were unable to complete all 4 cycles of of the patients also received postoperative radiotherapy
chemotherapy, with patient refusal and excessive (27.7% in the control group vs. 22.3% in the chemo-
toxicity being the most common reasons. A treatment therapy group). The primary endpoint was OS, with sec-
effect, thus, may have been harder to establish. The au- ondary endpoints being disease-free survival (DFS),
thors concluded that there was no evidence of superior- second primary cancers, and adverse effects. After a me-
ity of either regimen and adjuvant chemotherapy dian follow-up of 56 months, 44.5% of patients in the
should be restricted to clinical trials.6 chemotherapy arm versus 40.4% in the observation arm
In 2003, the results of the Adjuvant Lung Project were alive at 5 years (HR for death 0.86, 95% CI
Italy (ALPI) study were published.7 Between 1994 and 0.76e0.98, P < .03). In terms of DFS rate, 39.4% of pa-
1999, 1209 patients with stage I, II, or IIIA resected tients in the chemotherapy arm versus 34.3% in the
NSCLC were randomized to chemotherapy with 3 cy- observation arm were disease free at 5 years (HR 0.83,
cles of mitomycin C, vindesine, and cisplatin (MVP) 95% CI 0.74e0.94, P < .003). Higher than previous tri-
versus no treatment. The primary endpoint was OS, als, 73.4% of patients in the chemotherapy arm were
with secondary endpoints being progression-free sur- compliant with the planned cumulative dosage of
vival (PFS) and toxicity. The results showed no statisti- cisplatin. The absolute 5-year benefit in survival was
cally significant OS difference between the two groups 4.1%, consistent with that predicted by the 1995 Non-
after a follow-up of 64.5 months (HR 0.96, 95% CI small Cell Lung Cancer Collaborative Group metaanal-
0.81e1.13, P ¼ .589). Similarly, there was no difference ysis. Subgroup differences between treatment and stage
in PFS (HR ¼ 0.89, 95% CI 0.76e1.03, P ¼ .128). showed that patients receiving etoposide with cisplatin
However, similar to the ECOG 3590 study, only 69% and those with stage III disease appeared to benefit
of patients received the full 3 cycles of MVP, thus most.9 Long-term follow-up of this trial, however,
poor compliance and toxicity with the treatment showed that patients treated with an adjuvant
regimen were a drawback of the study and likely cisplatin-based regimen had a nonstatistically signifi-
contributed to the results.7 cant absolute survival benefit at 5 years of 3.9% (HR
The European Big Lung Trial, published in 2004, was 0.91, 95% CI 0.81e1.02, P ¼ .1), whereas the DFS ef-
a third negative randomized trial.8 A total of 381 pa- fect persisted with a significant absolute benefit of
tients with stage I, II, or III NSCLC were randomized 4.3% at 5 years (HR 0.88, 95% CI 0.78e0.98,
to either 3 cycles of weekly chemotherapy with 4 sepa- P ¼ .02).10
rate cisplatin-based chemotherapy regimens (cisplatin/ After the IALT trial results, both the National Cancer
vindesine, mitomycin/ifosfamide/cisplatin, mito- Institute of Canada (NCIC) JBR.10 and the Adjuvant
mycin/vinblastine/cisplatin, or vinorelbine/cisplatin) Navelbine International Trialist Association (ANITA)
or observation. Ninety-five percent of patients had a trial were able to also demonstrate a positive effect of
prior complete surgical resection. OS was the primary adjuvant cisplatin-based chemotherapy. The JBR.10
endpoint. Owing to toxicity, only 60% of patients in trial, published in 2005, enrolled 482 patients, with
the chemotherapy arm were given all 3 cycles of treat- stage IB or II resected NSCLC, from the United States
ment without delay or modifications, and 30% of pa- and Canada and randomized them to adjuvant cisplatin
tients in the chemotherapy arm experienced grade 3 or plus vinorelbine for 4 cycles versus observation with a
4 toxicity. The study failed to show an OS benefit be- primary endpoint of OS and secondary endpoints of
tween the arms with a median survival of 33.9 months recurrence-free survival (RFS), safety, toxicity, and qual-
in the chemotherapy arm and 32.6 months in the obser- ity of life.11 Contrary to previous studies, no patients
vation arm (HR 1.02, 95% CI 0.77e1.35, P ¼ .90).8 received any radiation during the course of their
The first large randomized positive adjuvant trial was therapy. After a median follow-up of 5.1 years (range
the International Adjuvant Lung Cancer Trial (IALT), 1.5e9.3 years) in the chemotherapy group and
CHAPTER 3 Adjuvant and Neoadjuvant Therapy in NoneSmall-Cell Lung Cancer 37

5.3 years (range 0.4e9 years) in the observation group, 62% of patients in the chemotherapy arm alive versus
the results showed an OS benefit of 94 months in the 64% in the control group (HR 1.10, 95% CI
chemotherapy arm versus 73 months in the observation 0.76e1.57). However, the test used for survival interac-
arm (HR 0.69, 95% CI 0.52e0.91, P ¼ .009). Five-year tion between tumor stage and chemotherapy was not
survival rates were 69% and 54% in the chemotherapy significant (P ¼ .07). With regard to radiation, the deci-
arm and observation arm, respectively, (P ¼ .03). The sion to give radiation was not randomized, even though
median RFS was not reached in the chemotherapy patients with N2 disease had a survival benefit in the
group versus 46.7 months in the observation group subgroup descriptive analysis. Adverse events in the
(HR 0.60, 95% CI 0.45e0.79, P < .001). Subgroup chemotherapy arm were similar to those reported previ-
analysis showed that survival advantage was limited ously using vinorelbine, although 2% patients died
to patients with stage II disease with a 20% 5-year sur- from treatment toxicity, possibly explained by higher
vival benefit (HR 0.59, 95% CI 0.42e0.85, P ¼ .004) doses used of cisplatin and vinorelbine. The authors
versus 7% in those with stage IB disease (P ¼ .79), concluded a positive survival difference of adjuvant
although this represented a smaller number of patients. chemotherapy, specifically in those patients with stage
Furthermore, those with RAS mutations also did not II and IIIA disease deriving the most benefit from adju-
appear to benefit from adjuvant chemotherapy (HR vant chemotherapy.13
0.95, 95% CI 0.53 to 1.71, P ¼ .87).11 Given both the positive and negative OS results of
The long-term follow-up of JBR.10 published in the aforementioned phase III trials, the Lung Adjuvant
2010, showed that OS persisted after a median follow- Cisplatin Evaluation (LACE) trial was a metaanalysis
up of 9.3 years (HR 0.78, 95% CI 0.61e0.99, P ¼ .04), designed to summate these overall effects, specifically
with an absolute survival benefit at 5 years of 11%.12 identifying treatment options and groups of patients
The benefit again appeared to be most pronounced in most likely to benefit from adjuvant chemotherapy.1
those patients with stage II disease. As a whole, patients The LACE analysis pooled the results of the five largest
with stage IB disease did not have a statistically signifi- trials in completely resected patients conducted after
cant benefit from chemotherapy (HR 1.03, 95% CI 0.7 the 1995 NSCLC metaanalysis. This included 4584 pa-
to 1.52, P ¼ 0.87); however, when stratified according tients from the ALPI, European Big Lung, IALT, JBR.10,
to tumors greater than 4 cm versus those less than and ANITA trials. Published in 2008, the results
4 cm, there was a trend toward improved survival in pa- confirmed a positive survival benefit of cisplatin-based
tients with larger tumors receiving adjuvant chemo- adjuvant chemotherapy, with a 5-year absolute benefit
therapy (HR 0.66, 95% CI 0.39e1.14, P ¼ .113).12 of 5.4% after a median follow-up of 5.2 years (HR
Similar to JBR.10, the ANITA trial was a phase III 0.89, 95% CI 0.82e0.96, P ¼ .005). DFS was also
study that randomized NSCLC patients to 4 cycles of favored in those who received adjuvant chemotherapy
adjuvant cisplatin plus vinorelbine or observation after with an absolute benefit of 5.8% at 5 years (HR 0.84,
complete surgical resection.13 This was an international 95% CI 0.78e0.91, P ¼ .001).1
study from 14 countries with 840 patients randomized When assessed by stage, both patients with stage IA
with stage IB (36%), II (24%), and IIIA (39%) disease. (HR 1.41, 95% CI 0.96e2.09) and IB (HR 0.93, 95%
Postoperative radiotherapy was optional and left to CI 0.78e1.10) disease did not derive benefit from adju-
each participating center to decide, although frequently vant chemotherapy, whereas those with node-positive
recommended to patients with node-positive disease. or stage II-IIIA disease had a significant positive survival
OS was the primary endpoint, with secondary end- benefit (HR 0.83, 95% CI 0.73e0.95). Comparisons
points being DFS and safety. After a median follow-up among the different cisplatin-based chemotherapy reg-
of 76 months, median OS in the chemotherapy arm imens (cisplatin plus vinorelbine, cisplatin plus etopo-
was 65.7 and 43.7 months in the observation arm side or vinca alkaloid, or cisplatin plus other) overall
(HR 0.80, 95% CI 0.66e0.96, P ¼ .017). The absolute showed no difference (P ¼ .11), although there was a
OS benefit at 5 years with chemotherapy was 8.6% trend towards improved OS with cisplatin plus vinorel-
which was also still evident at 7 years (8.4%). Median bine (HR 0.80, 95% CI 0.70e0.91), but these patients
DFS was 36.3 months in the chemotherapy arm versus also received higher doses of cisplatin in comparison.
20.7 months in the observation group (HR 0.76, 95% There was no change of chemotherapy effect with
CI 0.64e0.91, P ¼ .002). At 5 years, the absolute DFS planned or given radiotherapy for either OS or DFS.1
benefit of chemotherapy was 8.7%.13 The authors concluded that cisplatin-based chemo-
Similar to the JBR.10 trial, in patients with stage IB therapy is of benefit in patients with completely
disease there was no difference in 5-year survival, with resected early-stage NSCLC.
38 Pulmonary Adenocarcinoma: Approaches to Treatment

In 2011, a similar conclusion of survival benefit from (T2aN0), however, the NCCN guidelines recommend
adjuvant chemotherapy was made, based on the results chemotherapy for patients with high-risk disease after
of the Non-small Cell Lung Cancer Collaborative Group a complete or R0 resection (category 2A recommenda-
postoperative chemotherapy metaanalysis.14 There were tion). High-risk features include poorly differentiated
two metaanalyses presented. With the primary endpoint tumors, use of wedge resection, vascular invasion or
being OS, the first metaanalysis assessed 8447 patients visceral pleural involvement, unknown node status, or
from 34 randomized trials starting on or after Jan. 1, tumors greater than 4 cm.2 Data supporting tumor
1965 comparing surgery alone to surgery plus adjuvant size greater than 4 cm came initially from the Cancer
chemotherapy. The second metaanalysis assessed 2660 and Leukemia Group B (CALGB) 9633 trial, published
patients from 13 trials comparing surgery plus adjuvant in 2008.15 In this trial, 344 patients with T2N0 NSCLC
chemotherapy and radiotherapy versus surgery plus with complete resection were randomized to 4 cycles of
adjuvant radiotherapy. In the first metaanalysis, an ab- carboplatin plus paclitaxel versus observation, with OS
solute survival increase of 4% at 5 years was found being the primary endpoint. After a median follow-up
(from 60% to 64%) when chemotherapy was given after of 74 months the results showed that survival was not
surgery (HR 0.86, 95% CI 0.81e0.92, P < .0001). The statistically different between the two groups, with a
same survival benefit of 4% was also found (from median OS of 95 months in the chemotherapy arm
29% to 33%) in the second metaanalysis when chemo- and 78 months in the observation arm (HR 0.83, 95%
therapy was added to radiotherapy after surgery (HR CI 0.64e1.08, P ¼ .12). In the exploratory analysis,
0.88, 95% CI 0.81e0.97, P ¼ .009). The lower survival however, when patients were divided into those with
rates in the second metaanalysis were thought attribut- tumors greater than 4 cm in diameter and less than
able to inclusion of predominately stage III patients 4 cm, they found that patients with tumors greater
with a higher rate of incomplete resection.14 than 4 cm had a statistically significant survival differ-
In these metaanalyses, there was no difference ence in favor of adjuvant chemotherapy over observa-
among chemotherapy regimens, although the most reli- tion, with a 31% reduction in the risk of death and a
able evidence of benefit came from trials using a combi- median survival of 99 months versus 77 months,
nation of cisplatin plus vinorelbine in comparison to respectively (HR 0.69, 95% CI 0.48e0.99, P ¼ .043).15
the use of older vinca alkaloids (vinblastine, vindesine, In the Japan Lung Cancer Research Group (JLCRG)
and vincristine) or etoposide. The use of tegafur and study, 979 patients with resected stage IA and IB NSCLC
uracil or tegafur alone in older Asian studies appeared were randomized to adjuvant oral uracil and tegafur
to result in similar benefit to platinum-based regimens, (UFT) for 2 years or observation with the primary
even though it is difficult to apply these data to non- endpoint of OS.16 After a median follow-up of
Asian populations.14 Interestingly, there was no differ- 72 months, the results showed that the 5-year OS was
ence in the effect of platinum-based chemotherapy by 88% in the UFT group versus 85% in the control group,
stage in contrast to previous studies showing a trend to- a statistically significant difference (HR 0.71, 95% CI
ward poorer survival in patients getting adjuvant 0.52e0.98, P ¼ .04). This difference was driven by the
chemotherapy with stage IA disease and no significant 27% of patients with stage IB disease where the sub-
difference in those with stage IB disease.1 group analysis showed a significant survival benefit in
this group (HR 0.48, 95% CI 0.29e0.81, P ¼ .005)
versus no survival benefit in the larger subset of patients
STAGE I DISEASE with stage IA disease (HR 0.97, 95% CI 0.64e1.46,
Based on the LACE and Non-small Cell Lung Cancer P ¼ .87). In addition, they found that patients with tu-
Collaborative Group metaanalysis, adjuvant chemo- mors greater than 3 cm in size also derived a significant
therapy remains a recommended postsurgical approach survival benefit in contrast to those less than 3 cm.16
for patients with stage II and IIIA disease, with around a Based on these data, UFT remains an adjuvant treat-
5% OS benefit. The decision to treat patients with adju- ment option in Japan, although is not available in
vant chemotherapy for stage I disease is less concrete. Europe and North America.
Based on the aforementioned data, and the likely detri- The results of a smaller third randomized trial in
mental effect of adjuvant chemotherapy on patients Italy published in 2006 also supported the use of adju-
with stage IA (T1N0) disease,1 it is not recommended vant chemotherapy in patients with stage IB disease.17
that these patients receive adjuvant chemotherapy and Here 140 patients with stage IB NSCLC, after surgical
should proceed with standard surveillance after a com- resection, were randomized to observation or 6 cycles
plete surgical resection. In patients with stage IB disease of chemotherapy with cisplatin and etoposide, with
CHAPTER 3 Adjuvant and Neoadjuvant Therapy in NoneSmall-Cell Lung Cancer 39

the primary endpoint being OS and secondary endpoint as an abstract in 2016, showed that there was no differ-
being DFS. The results showed a significant OS differ- ence in survival among those patients who got bevaci-
ence between the groups, with a median OS of zumab and those who did not. There was also no
84.8 months in the chemotherapy arm versus difference in DFS. Furthermore, there was no OS or
41.6 months in the control arm (P ¼ .02). The 5-year DFS difference detected among the 4 different doublet
OS rate was 62% versus 42%, with the 10-year OS combinations in both squamous and nonsquamous
rate of 44% versus 20% in the chemotherapy and obser- histology. With regard to tolerance, in those patients
vation arms, respectively. DFS was also significant, with with nonsquamous histology administered with peme-
a median difference of 78.4 months in the chemo- trexed, it was associated with less grade 3e5 toxicity in
therapy arm versus 25.6 months in the observation comparison to other regimens. The cisplatin plus peme-
arm (P ¼ .0001).17 trexed regimen was also preferred among practitioners
Taken together, these three randomized trials suggest when given the opportunity to choose.18
that the subset of stage I patients with larger tumors A small phase II trial also showed improved tolera-
appear to benefit most from adjuvant chemotherapy. bility with pemetrexed as a pairing agent with cisplatin
The subgroup analyses of OS benefit in CALGB 9633 in early-stage resected NSCLC. The TREAT study, pub-
and JBR.10 in patients with stage IB tumors greater lished in 2013, randomized 132 patients with
than 4 cm, and in the JLCRG study of patients with tu- completely resected stage IB to IIIA NSCLC to 4 cycles
mors greater than 3 cm, lend support to this 11,15,16. In of adjuvant cisplatin plus pemetrexed or 4 cycles of
clinical practice, a 4-cm cutoff of when to consider adju- cisplatin plus vinorelbine.19 Forty-three percent had
vant chemotherapy in stage IB disease is often used. squamous histology and 57%, nonsquamous histology.
Other factors should be taken into account if the tumor The primary objective was clinical feasibility rate
is less than 4 cm as few reproducible and internation- defined as no grade 4 neutropenia or thrombocyto-
ally randomized data exist to support the use of adju- penia, no thrombocytopenia with bleeding, no grade
vant therapy otherwise in this patient population. The 3 or 4 febrile neutropenia or nonhematologic toxicity,
major aforementioned adjuvant phase III trials are and no premature withdrawal or death. Feasibility rates
reviewed in Table 3.1. were 95.5% in the cisplatin plus pemetrexed arm versus
75.4% in the cisplatin plus vinorelbine arm (P ¼ .001).
There was also significantly less hematological grade 3
CHOICE OF CHEMOTHERAPY or 4 toxicity in the cisplatin plus pemetrexed arm
It is generally agreed on that in North America and (P < .001), as well as greater delivery of total mean
Europe, the use of a cisplatin-based doublet regimen doses of drug given (P < .0001). To answer the question
is the standard of care when giving adjuvant chemo- of which regimen is superior, the JIPANG study is an
therapy. Previously mentioned studies have varied ongoing randomized phase III trial in Japan comparing
greatly in not only the dosage of cisplatin used in their cisplatin plus vinorelbine with cisplatin plus peme-
trials but also what chemotherapy agent cisplatin has trexed in completed resected nonsquamous NSCLC,
been paired with. In the LACE metaanalysis there was with OS being the primary endpoint.20
a trend toward improved OS with cisplatin plus vinor- With regard to the use of cisplatin or carboplatin, in
elbine, and this difference was significant when the a retrospective comparison of adjuvant carboplatin plus
other combinations in the study were pooled paclitaxel to cisplatin plus vinorelbine, 438 patients
(P ¼ .04).1 However, it still remained unclear what with completely resected NSCLC were evaluated.21 Re-
the optimal regimen was, and no conclusion could be sults showed that median OS was not different between
drawn regarding the effect of the various cisplatin doses the two groups with 5-year OS rates of 73% in those
and OS differences. who received carboplatin plus paclitaxel and 71% in
The E1505 trial was a randomized trial assessing the those who received cisplatin plus vinorelbine
effect of the addition of bevacizumab to a cisplatin- (P ¼ .71). There was also no difference in RFS
based doublet on 1501 patients with completely (P ¼ .68). The two different regimens brought different
resected NSCLC.18 Investigators could choose from 4 toxicities, with myalgias, sensory neuropathy, and alo-
chemotherapy options to pair with cisplatin: vinorel- pecia more common in the carboplatin plus paclitaxel
bine, gemcitabine, and docetaxel in those patients group versus vomiting, anemia, neutropenia, and fa-
with squamous histology, or any of these agents or tigue more common in the cisplatin plus vinorelbine
pemetrexed for patients with nonsquamous histology. group. There was also more discontinuation of treat-
The primary endpoint was OS. Results, first presented ment and dose reduction in the cisplatin plus
40 Pulmonary Adenocarcinoma: Approaches to Treatment

TABLE 3.1
Phase III Adjuvant Trials
Included Adjuvant Radiation Primary
Trial Stage Comparison Given N Endpoint Result P-value
6
ECOG 3590 II and IIIA Concurrent Yes 488 OS No significant 0.56
cisplatin/etoposide difference in survival
x4 cycles þ XRT vs.
XRT alone
ALPI7 I, II, and Mitomycin Optional 1209 OS 5-year 1% OS 0.589
IIIA C/vindesine/ benefit in the chemo
cisplatin x3 cycles arm
vs. observation
European Big I, II, and III Cisplatin þ either Optional 381 OS 2-year OS rate of 0.90
Lung Trial8 vindesine, 58% in the chemo
mitomycin/ arm versus 60% in
ifosfamide, the obs arm
mitocycin/
vinblastine or
vinorelbine x3 cycles
vs. observation
IALT9 I, II, and III Cisplatin þ either Optional 1867 OS 5-year OS rate of <0.03
etoposide, 44.5% in the chemo
vinorelbine, arm versus 40.4% in
vinblastine or the obs arm
vindesine x3-4
cycles vs.
observation
JBR.1011 IB and II Cisplatin/vinorelbine No 482 OS 5-year OS rate of 0.03
x4 cycles vs. 69% in the chemo
observation arm versus 54% in
the obs arm
ANITA13 IB, II, and Cisplatin/vinorelbine Optional 840 OS 5-year 8.6% OS 0.017
IIIA x4 cycles vs. benefit in the chemo
observation arm
CALGB IB Carboplatin/ No 344 OS 5-year OS rate of 0.125
963315 paclitaxel x4 cycles 60% in the chemo
vs. observation arm versus 58% in
the obs arm
JLCRG16 IA and IB Uracil/tegafur x2 No 979 OS 5-year OS rate of 0.04
years vs. 88% in the chemo
observation arm versus 85% in
the obs arm

ALPI, Adjuvant Lung Project Italy; ANITA, Adjuvant Navelbine International Trialist Association; chemo, chemotherapy; IALT, International
Adjuvant Lung Cancer Trial; JLCRG, Japan Lung Cancer Research Group; N, number of patients enrolled; obs, observation; versus, versus; OS,
overall survival; XRT, radiation therapy.

vinorelbine arm. The authors concluded that despite remains the preferred platinum agent of choice in the
this not being prospective data, carboplatin plus pacli- adjuvant setting. The CALGB 9633 trial is the only ran-
taxel can be considered as an effective and well tolerated domized reported study using carboplatin, with long-
adjuvant therapy regimen. term follow-up not confirming an OS benefit in the
Although retrospective data suggest potential equiv- stage IB patient population as a whole.15 Carboplatin
alency between cisplatin and carboplatin, cisplatin may be considered on an individual basis, specifically
Another random document with
no related content on Scribd:
CHOLELITHIASIS. BILIARY CALCULI. GALL
STONES.

In ruminants, omnivora, and carnivora, less often in horse, 1 to 1000, or more,


biliary sand to marble or more, casts; nucleus, bile pigment, blood mucus, etc.,
with concentric layers of cholesterine, pigment, salts and lime. Causes: idleness,
overfeeding, dry feeding, thickening of bile, colloids, bacteria, wintering in stable.
Colloids induce globular deposits. Parasites. Inflamed biliary epithelium secretes
excess of cholesterine and lime. Age. Acid bile, constipation. Diseased liver. Lack of
bile salts. In solipeds: all sizes and numbers, green, brown, yellow or white. Liver
disease. Symptoms: colic with depression, prostration, yellow or red urine, icterus,
sudden recovery. Treatment: sulphate of soda, olive oil, antispasmodics, warm
drinks, fomentations, salicylate of soda, chloroform, pasture, or succulent food,
and open air exercise. In cattle: musk odor; calculi dark green, yellowish green,
whitish or orange, dry red, all sizes and numbers. Causes: dry winter feeding, etc.
Lesions: dilated biliary ducts and bladder. catarrh, thickening, rupture, septic
peritonitis. Symptoms: relapsing colic, with icterus, prostration, and tender right
hypochondrium, in stalled animal on dry feeding. Suggestive: not pathognomonic.
Treatment: as in horse: succulent spring pasturage. In sheep: rare, musky odor;
concretions and casts common in distomatosis. Hypertrophied ducts. Symptoms of
distomatosis. Treatment: for distomatosis and calculi. In swine: rare: circular. In
carnivora: round, dark green, pin’s head to hazel nut. Symptoms: colics,
constipation, emesis, icterus, tender right hypochondrium, concurrent catarrh of
bowels, heart disease, dyspnœa, sudden relief. Mode of relief. Treatment: olive oil,
bile, sodium sulphate, or salicylate, antispasmodics, alkalies, enemata,
fomentations. Laxative food, exercise, open air.

Gall stones are most frequent in animals having a gall bladder.


Some medical writers say they are formed in the gall bladder only,
but the soliped which has no gall bladder has in particular instances
furnished hundreds of gall stones. Yet the ox, dog, sheep and pig are
the common victims of biliary calculi among our domestic animals.
In these the calculi appear to be mostly deposited from the stagnant
bile in the gall bladder, yet concretions on the biliary ducts and
hollow casts inside the ducts are by no means uncommon.
A gall stone may be single, or they may be multiple up to hundreds
or even thousands, and when very numerous they are individually
small, perhaps no larger than a pin’s head. They may, however,
attain the size of a marble or more, and by mutual pressure and wear
they assume various polygonal forms. If they lie apart in the gall
ducts or bladder they are regularly rounded. They are sometimes
mulberry shaped as if conglomerate. In other cases the solid masses
are so small as to have secured them the name of biliary sand. Casts
and incrustations in the ducts are not necessarily made up of smaller
globular masses.
On section a calculus shows a nucleus, composed of bile pigment,
blood, mucus, with the debris of parasites or bacteria. Around this
nucleus the calculus is deposited in concentric layers, of a hard
material consisting largely of cholesterine, but containing also bile
coloring matter, bile salts, and lime, in short all the constituents of
bile.
Causes. Various conditions contribute to the precipitation of
biliary solids in the form of calculi or encrustations. The most
prominent causes are: lack of exercise, overfeeding, dry feeding,
concentration of the bile, the presence of colloids and bacterian
infection.
Idleness is especially operative in cattle, which are quite subject to
biliary calculi and concretions, when shut up in the stall on
abundant, dry feeding for a long winter. They are not noticed in
stalled animals, that are fed watery or succulent rations, such as
green fodder, distiller’s or brewer’s swill, ensilage, brewer’s grains,
mashes, roots, potatoes, apples, pumpkins, and in case a tendency to
their formation is developed on the dry feeding of winter, the
concretions may be re-dissolved and entirely removed by the
succulent spring grass. A similar influence is noticed in the human
family, as the female sex living mostly indoors, and males pursuing
sedentary occupations furnish the greatest number of gall stones.
Concentration of bile results in part from muscular inactivity and
hepatic torpor, but also from overfeeding which loads the portal
blood and indirectly the bile with an excess of solids, and from dry
feeding which lessening the secretion of water leaves the bile more
dense and predisposed to precipitate its solids. The density of the
liquid, however, developed from a rich and dry ration and a
prolonged inactivity, may continue for a length of time, without the
occurrence of actual precipitation. It usually requires some
additional factor to make this predisposition a direct cause.
Presence of Colloids. This may be found in the presence of
solid or semi-solid particles. Just as the introduction of a thread into
a concentrated solution of sugar or salt will induce an instant
crystallization on the filament so the presence of solid bodies
determines a similar condensation in solid form of the solids of the
bile. But this tendency is increased materially if the solid body is
itself of a colloid or non-crystallizable material. Rainey and Ord have
shown experimentally that colloid bodies like mucus, albumen, pus,
blood, epithelial cells, not only determine the precipitation of
crystallizable salts from a strong solution, but that they cause the
precipitate to assume the form of globular or spherical particles,
which by gradual accretions on their surfaces tend to grow into
calculi. They found that salts which are deposited by mere chemical
reaction, without the intervention of colloids, appear in the form of
sharply defined angular crystals. The very fact that a precipitate
assumes a spherical form suggests the presence of colloids as an
active factor in the precipitation. Heat appears to intensify this
action, though probably the normal body temperature operates
mainly through the more active proliferation of bacteria.
Bacteridian infection. In connection with the action of colloids it
has been observed that when such bodies are in a condition of
fermentation they are much more potent as precipitants than if
inactive and sterile. But as all fermentations are the work of
microörganisms we are at once brought to the conclusion that
bacterial infection is one of the most potent causes of calculous
formations. The invading microbes operate upon the dissolved
solids, causing changes in their condition which reduce their
solubility, and thus determine the separation of calculi and
concretions in a manner allied to the precipitation of nitrates in the
soils.
But the same microbes operate in producing the colloids which
coöperate so effectively in the formation of calculi. The catarrhal
biliary ducts, or bladder, shed their epithelium, and transude white
and red globules, and form pus and an excess of mucus, all tending
to the separation of the biliary solids or forming nuclei on which
these solids may condense. The calculi and concretions tend in their
turn to maintain and advance the inflammation.
The access of the microbes to the biliary duct or bladder may be
effected through the blood of the portal vein or hepatic artery, or in
the new-born, through the umbilical vein from an infected navel. As
other modes of access may be named, a gradual advance from the
duodenum through the common bile duct, or more speedily on or in
the bodies of parasites (ascaris, strongylus, stephanurus, tænia,
echinococcus, distoma, fasciola, coccidia), etc.
Changes in the chemical composition of the bile have been
invoked as a cause of gall-stones, and Naunyn has found that the
inflamed biliary epithelium secretes an excess of cholesterine and
salts of lime. Thomas has also observed a great increase of
cholesterine in connection with a catarrhal angiocholitis in the dog.
Among other alleged causes of biliary calculi are advanced age
(Rigot, Hering), acidity of the bile (Zundel), constipation, and any
organic disease of the liver and bile ducts which interferes with
excretion of bile.
Age is supposed to act by inactivity, lessened secretion, hepatic
torpor, and the greater presumption of liver disease, acidity by the
precipitation of cholesterine and the dissolving of lime present in the
tissues, and constipation through hepatic inactivity, obstruction of
the flow of bile, and the tendency to infection through intestinal
fermentations.
It may be added that any diminution of glycocholate or
taurocholate of soda or potash, decreases the solubility of
cholesterine and bile acids and favors their precipitation.
GALL STONES IN SOLIPEDS.
Characters. The biliary calculi of solipeds are of all sizes and
shapes. When numerous they are mostly the size of a pin’s head
(Lucet). Birnbaum found in one animal 400 like peas. Dieckerhoff
has repeatedly found four or five of the size of a hazelnut. Verheyen
found one as large as an apple and says one exists at the Berlin
Veterinary College which weighs several pounds. Rigot found 90 in
the biliary ducts of an old horse, and Zundel records the death of a
stallion of twenty-six years from multiple gall stones.
The calculi may be little larger than grains of coarse sand. When
larger and solitary they are mostly globular or mulberry shaped; if
many are together they have become polygonal by friction. In other
cases notably with distomata they form hollow tubular incrustations
on the bile ducts, and contain a thick grumous bile. They are usually
of a green color, but may be yellowish brown, yellow, or whitish.
When cut across they present a nucleus enclosed in successive layers,
each successive one often differing from the last in color. Their
specific gravity is low, some will even float in water when taken from
the ducts, and all float when dried. Their composition is variable but
chiefly cholesterine, bile acids, resin and pigments, an albumoid
matter, with lime salts, etc. The nucleus may be the remains of a
dead parasite, epithelial cells, blood, pus, mucus, etc. The outer
layers are usually the hardest.
There may be attendant hypertrophy of the liver, cirrhosis,
amyloid and other degenerations, catarrh of the biliary ducts, and
distension or (according to Birnbaum) rupture of the portal vein.
Symptoms. There are no reliable diagnostic symptoms apart from
the colics which accompany the obstruction of a bile duct by a
passing calculus. These in the main resemble the colic of ordinary
indigestion, but they may be complicated by unusual depression and
nervous prostration. There may be drooping of the head, ears and
eyelids, watery eyes, resting the head on the manger or pushing it
against the wall. The urine is liable to be red or reddish (Jobelot),
and if it or the mucosæ show a yellowish tinge it is strongly
suggestive. There may be constipation or diarrhœa. The colics are
severe and may last for several days (Seaman, Lucet, Burgoin)
without fever and recover abruptly when the stone passes into the
duodenum. They recur, however, with the impaction of another
stone, and this intermittent feature, with the marked prostration,
and the access of slight jaundice with each colic furnishes the best
means of diagnosis.
Treatment. During the access of colic give a full dose (1 to 2 lbs.)
sulphate of soda in warm water along with some active
antispasmodic (belladonna, lobelia, chloroform, ether), and foment
the loins and hypochondriac regions. Olive oil in large doses (1 to 2
quarts) has been found effective. Salicylate of soda in full doses is
beneficial in stimulating the biliary secretion, diluting the bile, and
securing some measure of antisepsis in both bowels and liver.
In the intervals between the colics, sodium salicylate, sodium or
potassium carbonate, or olive oil in continuous doses may assist in
disintegrating the calculi or passing them on. Chloroform tends to
break them up by dissolving the cholesterin. But any such treatment
must be accompanied by the abundant ingestion of water, and this is
often best secured by a run in a rich green pasture. In the absence of
pasturage, succulent fruits, and roots, ensilage, mashes, and gruels
may be advantageously substituted, and conjoined with systematic
exercise in the open air.
GALL STONES IN CATTLE.
Characters. The biliary calculi of cattle are characterized by a faint
odor of musk, which becomes stronger on the addition of potassa
and the consequent disengagement of ammonia.
They are distinguished according to their color as dark green,
yellowish green, and orange, brownish or white. There are also the
sedimentary deposits (biliary sand).
The dark green calculi are the most frequent, and being found in
the gall bladder and larger bile ducts, they attain a larger size than
the others. Those in the gall bladder may be pear-shaped, and those
in the ducts, round, ovoid or cylindroid. They are often rough and
uneven on the surface with deep cracks and holes penetrating deeply
into their substance and often filled with cholesterin. The pigments
may change to a blood red when dried. The consistency of these
calculi varies, some being hard, resistant and heavy, while others are
soft and friable. All are composed of concentric layers around a
central nucleus as in those of the horse. They vary in weight up to
seven ounces in exceptional cases. They contain cholesterin, fat,
resin, pigments, and lime and magnesia salts.
The yellowish green calculi are usually spherical unless moulded
into polygonal shapes by mutual contact. In the last case they have
flattened surfaces. These have a firm consistency and are composed
of successive layers of nearly equal color and density surrounding the
central nucleus. They are on an average smaller than the dark green
variety but individual calculi have been found of three ounces.
The whitish or orange calculi are usually in the form of hollow
casts of the bile ducts having a dull white color externally and a
yellowish brown internally. They are usually thin, fragile and
crystalline and contain relatively more earthy salts than the two first
named varieties. From this cause also they have a higher specific
gravity. One specimen weighed 8 ounces.
The biliary sand or pulp, is made up of granules of a yellowish,
dark green or black color, forming with the bile a pultaceous mass
but drying into a consistent mass. It may be firmly adherent to the
mucosa of the gall bladder and require to be scraped off. These
granules may be looked on as the first step in the formation of calculi
or encrustations.
The causes of biliary calculi in cattle are mainly close confinement,
dry feeding, abundant rations, the presence of trematodes in the bile
ducts and finally microbian infection.
Lesions vary. With obstruction of the common bile duct or cystic
duct, there are usually dilatation of the bile ducts with fibrous
thickening of their walls so that they stand out as white branching
lines on the back of the liver. In extreme cases the common duct may
acquire the calibre of the small intestine. The gall bladder may
participate in this thickening (Chassaing) or may even rupture
(Proger, Shaw).
In connection with obstruction microbian infection extends
upward into the liver, and in rupture of the bladder an acute, diffuse
septic peritonitis follows (Chassaing). This only follows on infective
inflammation of the gall bladder. Aseptic bile causes little or no
irritation.
Symptoms. As in the horse, general symptoms of ill health or
hepatic disorder are not pathognomonic. The presence of
intermittent attacks of constipation, and colic, with icterus,
tympanies and violent efforts at expulsion are the diagnostic
symptoms of an acute attack. Pulsation and respiration are
accelerated, and the urine dense, high colored, oily and slightly
yellow. Reboul has noticed that symptoms are aggravated on
exposure to cold; there are great prostration and dullness, frequent
moaning and marked indications of tenderness when the right
hypochondrium is percussed. Charlot has observed that the only
symptoms may be persistent jaundice with scanty, high colored
urine, containing some sediment.
Treatment is essentially the same as in the horse. Vanswieten and
Verheyen draw special attention to the fact that whereas biliary
calculi are very common in cattle during winter, they are rarely
found in animals that have been for even a short period on the spring
grass. Spring pasture is therefore the best therapeutic agent. During
paroxysms of colic, Glauber salts, or olive oil, antispasmodics and
fomentations over the liver are to be tried. In the intervals salicylate
of soda, sodium and potassium carbonate, olive oil, chloroform, and
ether may be used. Abundance of water and aqueous rations are
essential.
GALL STONES IN SHEEP.
Calculi are very rare. One described by Morton had a brownish
yellow color on its surface, and a white color spotted with green
internally; it had a bitter taste, colored saliva yellow, and melted
when heated, diffusing the odor of musk. It weighed twelve grains
and contained 70 per cent. of cholesterin, calcic phosphate and
carbonate and the usual biliary elements.
But if spherical calculi are rare, concretions and casts of the bile
ducts are common, especially in distomatosis. These are of a
yellowish, reddish, greenish or blackish brown, and form granular
plates, or veritable cylindroid casts often firmly adherent to the
mucous membrane of the duct.
In such cases the walls of the encrusted ducts are hypertrophied
and stand out on the back of the liver as white bands diverging from
the portal fissure.
Apart from the usual symptoms of distomatosis no special
indications have been observed.
Treatment is primarily that for distomatosis, to which the general
measures advised for calculi may be added.
GALL STONES IN SWINE.

Characters. The calculi are spherical, rough or on their opposed


surfaces flat, clear and glistening where they have become polished
by friction. They are found as a fine sand or as calculi the largest of
which have been 75 grains, and of a high density (1303 to 1484).
Bruckmüller found that they contained carbonate of lime and biliary
mucus. Verheyen found biliary resin, mucus, pigment, and a little fat.
They are rare in fat hogs in America. No diagnostic symptoms have
been observed.
GALL STONES IN DOG AND CAT.
These are more or less spherical, dark brownish green, and usually
found in the gall bladder or larger bile ducts. They may vary in size
from a pea to a hazel nut. Their chemical analysis is wanting.
Symptoms. There may be evidence of biliary obstruction and if
this occurs intermittently and is associated with colic, it becomes
somewhat characteristic. Constipation, emesis, icterus, and
sometimes tenderness of the right hypochondrium would indicate
the source of the colic. A pre-existing and concurrent catarrh of the
bowels corroborates these indications.
Cadeac explains that the obstructing calculus is called on to resist
the impulse of the bile forced upon it by the spasmodic contraction of
the bile ducts, which distends the bile duct immediately back of the
stone to perhaps ten times its normal size. Then under a suspension
of the spasm or even an antiperistaltic contraction of the duct, the
calculus is forced back into the dilated portion or even into the gall-
bladder, and the attack is relieved. Under repeated irritations of this
kind the inflammation of the bile ducts extends into the liver and
determines cirrhosis. The irritation further through the sympathetic
produces a reflex constriction of the pulmonary capillaries, with the
natural results of increasing tension of the pulmonary artery and
right heart, and dilatation and degeneration of the walls of the latter
even in the best nourished animals. Thus dyspnœa and modified
heart sounds (murmurs) may be symptoms of biliary calculi.
Treatment. Three or four ounces of olive oil were found to greatly
increase the quantity and fluidity of the bile in from thirty to forty-
five minutes. Bile, sulphate of soda and salicylate of soda are
excellent cholagogues, and the latter at the same time an antiseptic.
Anti-spasmodics are especially indicated to relieve the colics, but
they must be used in relatively smaller doses than in the herbivora.
Potassic and sodic carbonates or tartrates (Vichy) may be used as
enemas if they cannot be administered by the mouth. Fomentations
may be resorted to. The food must be laxative and aqueous, and
exercise must be imposed as far as the animal can bear it.
FOREIGN BODIES IN THE LIVER.
In horse: spikes of leguminosæ, barley awns. Symptoms: of internal
hemorrhage, pallor, weakness, vertigo, death; jaundice, prostration, stupor,
weakness, crossing of fore limbs, tender right hypochondrium. In cattle: bodies
passing from rumen. In swine, sand. In dog, sharp bodies from stomach.
Treatment: laparotomy.
Foreign bodies are rare in the liver in our domestic animals.
Horse. St. Cyr has found the spikes of leguminosæ and Megnin the
beards of barley. St. Cyr believed that he traced the passage followed
by the stalk through the walls of the duodenum, and portal vein
where it divided to be distributed through the liver. At the point of
supposed entrance the walls of the vena portæ were thickened and its
lumen filled with clots. The further course of the portal vein and its
branches showed similar thickening and clots, and on the branch
leading to the right lobe was a large abscess containing 4 decilitres of
pus. Clots extended into the splenic, omental and mesenteric veins,
and between the folds of the mesentery of the small intestine were a
number of minute ruptures and blood extravasations.
Megnin found traces of the passage of the barley beards through
the gastric walls and into the substance of the liver close to the portal
fissure. Around the centre where the barbs were implanted there was
an irregular hæmorrhagic extravasation in the liver, and in the
abdominal cavity an effusion of 8 or 10 quarts of blood.
Symptoms. In such a case the only definite symptoms are those of
internal hemorrhage, pallor of the mucous membranes, gradually
increasing weakness, vertigo, unsteady gait, and an early death. In
more protracted cases slight jaundice, dullness, prostration, stupor,
drooping of head, ears and eyelids, resting it on the manger or walls,
muscular weakness, crossing of the front limbs, and it may be
tenderness on percussion on the right side of the chest posteriorly. It
resembles the coma or immobility of the horse but the patient backs
more easily.
Cattle. In ruminants sharp pointed bodies passing from the rumen
will occasionally penetrate the liver, and give rise to symptoms of
hepatic disorder. Augenheister found in a cow dilatation of the larger
bile ducts, which contained about 10 quarts of sand, that had
apparently entered from the duodenum by the common bile duct
which had an orifice of an inch in diameter.
Pig. The gall ducts of a pig’s liver, in the Veterinary College of
Berlin contains a large amount of sand (Gurlt).
Dog. The liver is exceptionally perforated by sharp pointed bodies
coming from the stomach. Cadeac and Blanc report three cases of
needle in the liver. Blanc’s case had been killed because of old age;
one of Cadeac’s showed symptoms resembling rabies.
Treatment of these cases would be very hopeless as nothing short
of laparotomy and the removal of the foreign body would promise
success.
TUMORS OF THE LIVER. NEW GROWTH.

Largely secondary, from stomach, intestine, lymph glands, spleen, pancreas; the
hepatic tumor may be disproportionately large. In horse: sarcoma rapidly growing
soft, succulent, slow-growing, fibrous, tough, stroma with round or spindle shaped
cells and nuclei. Symptoms: emaciation, icterus, enlarged liver, rounded tumors on
rectal examination. Melanoma, in old gray or white horses, with similar formations
elsewhere; not always malignant. Lymphadenoma. Angioma. Carcinoma.
Epithelioma, lesions, nodular masses, white or grayish on section, and having firm
stroma with alveoli filled with varied cells with refrangent, deeply staining, large,
multiple nuclei, cancerous cachexia and variable hepatic disorder. In cattle:
sarcoma, adenoma, angioma, cystoma, carcinoma, epithelioma. In sheep:
adenoma, carcinoma. In dog: lipoma, sarcoma, encephaloid, carcinoma,
epithelioma. Wasting and emaciation, yellowish pallor, temporal atrophy, ascites,
liver enlargement, tender right hypochondrium, dyspepsia, symptoms of primary
deposits elsewhere.

The great quantity of blood which passes through the liver lays it
open, in a very decided way, to the implantation of germs and
biological morbid products. Hence tumors of the liver are largely
secondary, the primary ones being found mostly in the stomach,
intestine, abdominal lymph glands, spleen and pancreas. The
primary neoplasm is often comparatively small, while the hepatic
one supplied with a great excess of blood may be by far the most
striking morbid lesion. The hepatic tumors are mostly of the nature
of angioma, sarcoma, melanoma, adenoma, lipoma, cystoma,
carcinoma, and epithelioma.
NEOPLASMS IN HORSES LIVER.
Sarcoma. This is usually a secondary formation from the primary
tumors in the spleen and peritoneum, and it occurs as multiple
masses throughout the substance of the gland. The liver is greatly
increased in size, extending far beyond the last rib on the right side,
and weighing when removed as high as 70 ℔s. (Mason), or even 88
℔s. (Cadeac), in extreme cases.
The whole surface of the liver may show bulging, rounded masses,
and the morbid growth may have involved the capsule and caused
adhesion to the back of the diaphragm (Bächstädt). The cut surface
of the neoplasm is smooth, elastic, yellowish and circular or oval in
outline. It may have a variable consistency—friable or tough,
according to the activity of growth and the relative abundance of cells
and stroma. The portal glands are hypertrophied and thrombosis of
the portal vein is not uncommon.
Microscopic examination of the dark red scrapings shows
numerous blood globules, intermixed with the round or spindle
shaped cells and nuclei of the tumor. Sections of the tumor show
these cells surrounded by a comparatively sparse fibrillated stroma.
The round cells may vary from .005 to .05 m.m. They contain one or
more rather large nuclei and a number of refrangent nucleoli. The
nuclei are often set free by the bursting of the cells in the scrapings.
They become much more clearly defined when treated with a weak
solution of acetic acid. Small grayish areas in the mass of the tumor
represent the original structure of the liver, the cells of which have
become swollen and fatty.
A liquid effusion more or less deeply tinged with red is usually
found in the abdominal cavity.
Symptoms are those of a wasting disease, with some icterus,
sometimes digestive disorder, and a marked enlargement of the liver.
The last feature can be easily diagnosed by palpation and percussion.
If an examination through the rectum detects the enlargement and
irregular rounded swellings of the surface of the liver or spleen, or
the existence of rounded tumors in the mesentery or sublumbar
region, this will be corroborative. The precise nature of the
neoplasms can only be ascertained after death.
Melanoma. Melanosis of the liver is comparatively frequent,
especially in gray horses, and above all when they are aging and
passing from dark gray to white. In many cases a more certain
diagnosis can be made than in sarcoma for the reason that primary
melanotic neoplasms are especially likely to occur on or near the
naturally dark portions of the skin, as beneath the tail, around the
anus or vulva, in the perineum, sheath, eyelids, axilla, etc. The extent
of the disease is likely to be striking, the liver, next to the spleen,
being the greatest internal centre for melanosis. The whole organ
may be infiltrated so that in the end its outer surface is completely
hidden by melanotic deposit. The surface deposits tend to project in
more or less rounded, smooth masses of varying size according to the
age of the deposit and the rapidity of its growth. Individual deposits
may vary in size from a pea to a mass of 40 or 50 lbs. They are
moderately firm, and resistant, and maintain a globular or ovoid
outline. The color of the melanotic deposits is a deep black with a
violet or bluish tint. If the pigmentary deposit is in its early stage it
may be of a dark gray. The deposits are firmer than the intervening
liver tissue and rarely soften or suppurate.
Melanosis in the horse is not always the malignant disease that it
shows itself to be in man, and extensive deposits may take place
externally and considerable formations in the liver and other internal
organs without serious impairment of the general health. It is only in
very advanced conditions of melanosis of the liver that appreciable
hepatic disorder is observed. If, however, there is marked
enlargement of the liver, in a white or gray horse, which shows
melanotic tumors on the surface, hepatic melanosis may be inferred.
Lymphadenoma. Adenoid Tumor. Lienaux describes cases of this
kind in which the liver was mottled by white points which presented
the microscopical character of adenoid tissue, cells enclosing a
follicle and a rich investing network of capillaries.
Angioma. These are rare in the horse’s liver, but have been
described by Blanc and Trasbot as multiple, spongy tumors on the
anterior of the middle lobe, and to a less extent in the right and left,
of a blackish brown color, soft and fluctuating. The largest mass was
the size of an apple, and on section they were found to be composed
of vascular or erectile tissue. The tendency is to rupture and
extensive extravasation of blood (30 to 40 lbs.) into the peritoneum.
Carcinoma. Epithelioma. These forms of malignant disease are
not uncommon in the liver as secondary deposits, the primary
lesions being found in the spleen, stomach, intestine, or pancreas, or
more distant still, in the lungs. The grafting or colonization of the
cancer in the liver depends on the transmission of its elements
through the vena portæ in the one case, and through the pulmonary
veins, the left heart and hepatic artery in the other.
Lesions. The liver may be greatly enlarged, weighing twenty-seven
pounds (Benjamin) to forty-three pounds (Chauveau), hard, firm,
and studded with firm nodules of varying sizes. These stand out from
the surface, giving an irregular nodular appearance, and are
scattered through its substance where, on section, they appear as
gray or white fibrous, resistant, spheroidal masses shading off to a
reddish tinge in their outer layers. Microscopically these consist of a
more or less abundant fibrous stroma, enclosing, communicating
alveoli filled with cells of various shapes and sizes, with large nuclei
(often multiple) which stain deeply in pigments. The relative amount
of fibrous stroma and cells determines the consistency of the mass,
and whether it approximates to the hard cancer or the soft. In the
horse’s liver they are usually hard, and, on scraping off the cut
surface, yield only a limited quantity of cancer juice. In the epithelial
form, which embraces nearly all that have originated from primary
malignant growth in the walls of the intestine, the epithelioid cells,
flattened, cubical, polyhedral, etc., are arranged in spheroidal masses
or cylindrical extensions, which infiltrate the tissues more or less.
These seem in some cases to commence in the radical bile ducts
(Martin), and in others in the minor coats of the larger biliary ducts.
As the disease advances a brownish liquid effusion is found in the
abdomen, and nodular masses formed on the surface of the
peritoneum.
Symptoms. As in other tumors of the liver these are obscure. As
the disease advances there may be œdema of the legs and sheath,
indications of ascites, stiff movements, icterus, occasional colics,
tympanies, and diarrhœa. Nervous symptoms may also appear, such
as dullness, stupor, coma, vertigo and spasms. Emaciation goes on
rapidly and death soon supervenes.
TUMORS OF THE LIVER IN CATTLE.

Sarcoma. Round or spindle shaped celled sarcomata have been


described by Sodero and Cadeac, leading in one case to perforation
of the vena portæ and death by hæmorrhage into the peritoneum.
Adenoma. Martin records a case of a hepatic tumor formed of
adenoid tissue which had extended into the vena portæ, and
microscopically presented a cylindroid character. In the advanced
stages it caused some jaundice, digestive disorder, obstinate
constipation, progressive and extreme emaciation, and weakness
which kept the animal constantly in a recumbent position.
Angioma. These are rarely seen in the young but are comparatively
common in old cows as they are in aged men. They form masses of a
dark red color and very variable size, and have a limiting sac of
connective tissue or merge into the adjacent hepatic structure.
Microscopically they consist of a series of irregular lacunæ filled with
liquid blood, blood clots, or leucocytes, and communicating with
small blood vessels in the walls and partitions. They are believed to
be formed by dilatation of the liver capillaries with subsequent
thickening of their distended walls, and atrophy of the nearest liver
cells. Cases of the kind have been recorded by Kitt, Martin,
McFadyean, Saake, Van der Sluys, Korevaar and others. Though
often seen in abattoirs, they seem to have little effect on the general
health, and no special symptoms have been noted as indicating their
existence.
Congenital Cysts. These are found on the anterior surface or lower
border of the liver in young calves. They have no connection with the
blood vessels, nor biliary canals, contain no head of larval tænia, and
do not constantly show the presence of any particular bacterium.
These walls are thin and their contents alkaline, with sometimes
slight blood extravasation, or a yellowish deposit in which cocci have

You might also like