Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

Big 2017 Change on PSA by USPSTF

Marc B Garnick, MD, Editor in Chief, HMS Annual Report on Prostate Diseases; Gorman Professor of Medicine, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA . E. David Crawford, MD, Professor of Urology and Radiation Oncology at The University of Colorado, Anschutz Campus, Aurora, CO; Medical Advisor and Founder, 3DBiopsy, Inc., Aurora, CO

Q: The new draft USPSTF recommendations for rapid comment on use of PSA for screening represent a big change from 2012. They now read: “The decision about whether to be screened for prostate cancer should be an individual one. The USPSTF recommends that clinicians inform men ages 55 to 69 years about the potential benefits and harms of prostate-specific antigen (PSA)–based screening for prostate cancer” and much more. Shared patient-physician decision making. What do you think?

A: The long awaited updated recommendations from the United States Preventive Services Task Force, issued at their traditional 5-year interval, got it right–Again! While the 2012 “D” recommendation from the Task Force was met with great skepticism and animosity from several groups, the change in the 2017 recommendation now emphasizes the importance of the Task Force’s continuing evaluations of new and more mature data.
In a series of four detailed, analytic and erudite publications that provide the underpinnings for their new recommendations, even the most serious students of prostate cancer and prostate cancer screening–ourselves included–can appreciate the enormous task that members of the Task Force and its sub-sections undertook. The result not only justifies the correct C recommendation for those men between ages of 55 and 69 and the unchanged D recommendation for those above 70. Moreover, the recommendation calling for the serious need for more study about the complexities facing men who are at increased risk for prostate cancer– African-American and those with a significant family history–should serve as a call to action for research agencies whose worldwide populations are affected by this common cancer. The complexities of prostate cancer biology are outlined in these accompanying publications and will undoubtedly provide avenues for productive future research programs.
The changes in the Task Force’s recommendations appropriately relied heavily on several key studies- among them, the PLCO, ERSPC and ProtecT. Of the key factors that have become available since issuance of the 2012 “D” guidelines were both the increasing utilization of active surveillance–to help address the issue of over-diagnosis and the harms of overtreatment–and the potential that the development of metastases could be decreased by treatment compared to active maintenance. This latter assessment emanated from the UK ProtecT study and its context must appreciate that many of the patients enrolled in that study who received no active treatment had cancer characteristics such as high Gleason scores that would generally have prompted interventions here in the United States.
In the end, the Task Force has in fact provided a strong rationale for what is commonly happening in practice today despite the universal 2012 “D” recommendations. Patients should consider shared decision making with their health provider about the harms and benefits of undergoing screening, and treatment if a cancer is found, and that values and preferences of the individual patient should play an important role. But, today, as was the case in 2012, the ability to show an overall survival benefit from any screening recommendation still eludes us and the cancer-specific survival benefit, if one exists at all, is at best, very modest. Hopefully, these updated screening guidelines will encourage meaningful research to enable real advances at the patient level to be achieved.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

Forget Moonshots: Biomedicine Needs an Air Traffic Control System

Jeff Shrager, PhD, Director of Research, Cancer Commons; Adjunct Professor, Symbolic Systems Program, Stanford University

Q: There never seem to be enough patients matched to cancer clinical trials to quickly test new cancer treatments. Might there be a better way, using new communication technology?

A: Among the few things that everyone can agree upon, one is surely that biomedicine is not an efficient engineering system — that is, a system that efficiently (in time, cost, lives, or whatever else you’d like to measure) reaches its goals to alleviate suffering, etc. When we think of efficient engineering systems, companies such as Apple, Boeing, Google, Tesla, NASA, GE, and Toyota come to mind. Perhaps Big Pharma and parts of the VA and military are efficient engineering systems but surely biomedicine, as a whole, is not.
Granted, biomedicine faces the highly complex problem of matching patients to treatments while at the same time creating and ethically testing novel treatments with budgets that are under constant pressure. But this problem is faced by a huge number of extremely smart people. What is the difficulty? I contend that the difficulty is, in large part, lack of coordination.
When I think of highly efficient coordination, I think of the Air Traffic Control system. The notable pain of air travel notwithstanding, one has to admit that the flight coordination is nearly magical! You can fly from New York to London in 5hrs for $500 while strapped into a 20x200ft aluminum tube (full of explosive fuel) traveling at 500mph, 50,000ft high in an airspace shared with 100,000 other flights doing the same thing in every direction all over the world. And you never think about dying! Well, you may think about it, but realistically you shouldn’t because air travel is spectacularly safe. Instead, worry about escalators and bicycles!
I contend that we can learn from the spectacular success of Air Traffic Control in order to improve biomedicine’s engineering efficiency by improving its coordination. Furthermore, I contend that we can do this without compromising ethics, whilst making everyone involved rich and happy–or at least happy that they are not pissing away money and lives.
Essentially, we have proposed to build a Biomedical Air Traffic Control System that will efficiently search the huge space of plausible treatment regimens, crossed with the relatively small number of available patients in ever diminishing cohorts. I call this approach Global Cumulative Treatment Analysis (GCTA), and we can build it on much more modern technology and with much better understanding of science and engineering than the aviation community had when they built theirs.
GCTA is described in the following video and in more detail in Theoretical Issues for Global Cumulative Treatment Analysis (GCTA).

Cancer Commons is putting together a series of virtual workshops to work out the practical details of GCTA. If you would like to be in the loop for these, please contact me at jshrager@stanford.edu.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Back to Full Blog List

Connect With Us

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

Proposed FDA “Conditional Approval”- More Details


Al Musella, DPM, President, Musella Foundation For Brain Tumor Research & Information, Inc., Hewlett, NY. Marty Tenenbaum, PhD, Founder and Chair, Cancer Commons, Los Altos, CA.

Q: Your April 5, 2017 blog post that proposed a new “Conditional” category for FDA drug approval elicited a number of positive and negative responses. Please explain the proposal in more detail to enable better reader understanding.
A: In Response to “Conditional Approval: Right Solution for the Wrong Problem” by
Shannon Brownlee:
We appreciate Ms. Brownlee’s comments on our recent blog post, but think she missed our key points—perhaps we weren’t clear enough:

  • Conditional approval isn’t intended for cancers that have other options—like some types of breast cancer—but rather for the invariably fatal cancers like glioblastoma multiforme (GBM), for which there aren’t any good FDA-approved options.
  • Conditional approval requires not just evidence of safety, but evidence that the drug has promise, at least in some patients.
  • Using the registry is just a different way of collecting the data that traditionally is collected in clinical trials. Instead of a traditional trial in which only highly selected patients participate and relatively few doctors collect the data, we will be collecting data on the entire range of patients from many doctors.

Having many doctors and patients participate removes a lot of the bias inherent in clinical trials. For example, consider a doctor using an experimental treatment on “thousands” of brain tumor patients in 100 clinical trials in which only a handful of doctors are collecting all of the data. All of the doctors involved have a major financial interest in the outcome. We still have no idea if this treatment helps or hurts—and it has been going on for 40 years. With our system, we would know in a year if it helps or hurts.
Moreover, Ms. Brownlee’s arguments extolling the virtues of randomized clinical trials (RCTs) may reflect an unfamiliarity with the cost-effectiveness of “registry trials,” especially when coupled with modern Bayesian designs.
With the traditional approach, we have been lucky to get 3 or 4 treatments approved in the last 40 years. With our approach, we can expect at least 3–6 new treatments conditionally approved each year, as the cost to get approval would be on the order of 1–2% of the cost using the current traditional approach.

  • As Ms. Brownlee states, “It took more than a decade to accumulate enough patients” in an RCT to demonstrate the ineffectiveness of autologous bone marrow transplant (ABMT) in breast cancer. Further, “A few patients may actually have been helped by the Avastin, but there is no way to predict ahead of time whether patients would be helped or harmed.” These points demonstrate the limitations of classical RCTs versus Bayesian point-of-care trials that can be run on top of a registry.
  • The big challenge and opportunity regarding investigational treatments that show promise in some patients is to identify and continuously refine the cohort for whom an intervention is effective, as efficiently as possible. This requires a Bayesian approach, which can rapidly replicate successes and discard failures, not accruing a large trial to test a specific hypothesis, which is likely to be wrong.
  • A registry that captures biomarkers, treatments, and outcomes from patients undergoing a variety of interventions can provide rigorous cross-controls, which are every bit as valid as those provided by randomization.

Using Ms. Brownlee’s example of ABMT, in our system, the registry would have quickly picked up that it was not as good as standard chemotherapy. The current system allowed 41,000 women to use a treatment whose effectiveness was unknown because nobody was tracking it. An RCT took 10 years to find an answer that our registry may have had in as little as 1 year, saving many lives.
Ms. Brownlee states that the registry can’t show efficacy. We disagree. If you have the majority of patients being tracked in the registry, you can use all of the patients who are NOT taking the treatment as the control group. Comparing to the old historic control is useless for brain cancer—almost everyone died and we did not collect the necessary biomic data to correctly match them with current patients.
The comparison that needs to be made is which of many possible new treatments and combinations works the best. Some patients would probably stick with the old standard of care—we would be encouraging them to participate as well—so we can see if the current standard is better than any of the new treatments.
We are talking about using conditional approval only in cases where there really is no acceptable standard of care. And we ARE looking for new drugs and combinations that have extraordinary power to improve outcomes, not looking for something that extends life by weeks. We agree it wouldn’t make sense in a disease where the standard treatments offer hope.
In summary, our proposal is not to minimize the research, it is to maximize the amount of research done to a drug, just in a different time period—after phase 1 instead of before the end of phase 3.
As to picking up idiosyncratic reactions, by definition, these are rare and do require a large group of patients being tracked to identify how frequently they occur. With our registry, it is simple to analyze the data and get an early warning. Having the genomic data would allow us to try to figure out which patients are most likely to have such a problem. With traditional trials, which usually only allow a select population to be tested, a reaction that only occurs in the elderly or in minorities—which are underrepresented— may never be found. Once a drug is approved and underrepresented groups use it, side effects are not tracked as closely as they would be in our registry.
For more reference, please read “Rapid Learning for Precision Oncology” published in Nature on Jan 21, 2014. We include a free PDF download. Please see below for a 3-minute video showing how the Bayesian approach can be applied to medical research in a new method called Global Cumulative Treatment Analysis (GCTA).


Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

Next Steps for Improving Clinical Evidence and Consistency of Payment for Precision Oncology

Kathryn A. Phillips, PhD, Professor of Health Economics and Health Services Research; Founding Director UCSF Center for Translational and Policy Research on Personalized Medicine; Dept. of Clinical Pharmacy; Institute for Health Policy Studies; Comprehensive Cancer Center
University of California, San Francisco

Q: Adoption and insurer coverage for precision oncology may require evidence that it can improve patient outcomes. The current situation is confusing for many. What are some practical next steps that workers in the field can take to improve clinical evidence and consistency of payment?

A: It’s been said that the greatest challenges facing genomic medicine are not scientific, but economic. Much has been written about the need for improved clinical evidence and consistent reimbursement policies, but there have been relatively few studies that illuminate what steps can be taken to address them. Two new studies from the University of California San Francisco Center for Translational and Policy Research on Personalized Medicine (TRANSPERS) address these questions.
A newly published study in Genetics in Medicine combines insights from TRANSPERS collaborators and leading genomic medicine experts to identify evidence gaps in genomic medicine that comparative effectiveness research can address, with direct relevance to precision oncology. For genomic/precision medicine to fulfill its potential, it must be (1) evidence-based and (2) consider a full range of patient outcomes. Does the literature to date suggest that these objectives have been met? TRANSPERS and experts from multiple institutions addressed this question. This is the first study that uses a systematic structured literature review (combined with expert input) to provide an overarching assessment of comparative effectiveness research for genomic medicine. We found that all included reviews (N=21) identified potentially important clinical applications of the genomic medicine interventions. Most had significant methodological weaknesses and there were few studies of conditions other than cancer. There were only a few analyses examining a broad range of patient-centered outcomes. Our findings provide next steps about where to focus future research activities and policy initiatives by identifying conditions, tests, and interventions where comparative effectiveness questions may be appropriate for study. We also discuss the limitations of prior research and how they could be addressed.
Specifically for precision oncology, we found that studies are needed to measure whether tumor sequencing tests lead to better clinical outcomes than alternative prognostic methods in different stages of common cancers. Additionally, studies are needed for cancer risk assessment panels that examine the consequences of testing for individuals and families including acceptability to patients, adherence to screening, delivery of genomic testing, and models to estimate the incremental net benefit of testing and optimal testing intervals.
Another recent publication from TRANSPERS identifies opportunities to resolve reimbursement challenges of genetic panel tests for cancer risk assessment. This study, published in the Journal of the National Comprehensive Cancer Network used data from payers themselves to address not only reimbursement challenges but also opportunities for resolving those challenges. Hereditary cancer panels – testing for multiple genes and syndromes – are rapidly transforming cancer risk assessment but are controversial and lack formal insurance coverage. Our study of private payers found a number of barriers to coverage for hereditary cancer panels including poor fit with coverage frameworks, insufficient evidence, and departure from pedigree/family history-based testing toward population-based genetic screening. Opportunities for addressing these challenges includes refining target populations, developing evidence of actionability and pathogenicity/penetrance, and creating infrastructure and standards for informing and re-contacting patients. We also need to separate research from clinical use in the hybrid clinical research setting and adjust coverage frameworks. Our findings have particular relevance to the NIH’s Precision Medicine Initiative, which will assemble and study an unprecedented cohort of one million or more volunteers who will contribute genomic, clinical and lifestyle data to accelerate genetic science.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

Conditional Approval: Right Solution for the Wrong Problem

Shannon Brownlee, MS, Senior Vice President of the Lown Institute, a think tank in Boston. She is also co-founder of the Right Care Alliance, a social movement for transforming health care.

Q: Musella and Tenenbaum recently proposed a new way, called conditional approval, for the American FDA to move potentially useful drugs to a patient market. They wrote that safety would be covered and efficacy assessed by a registry. What do you think of that idea?
A: Imagine if there were a way to speed up the discovery and testing of drugs for cancer. Al Musella and Marty Tenenbaum, founders of two cancer patient advocacy organizations, think they have just such a plan.
They have proposed giving new cancer drugs conditional approval, allowing them on the market after Phase I (safety) trials in as few as 50 patients. Patients and their doctors would be free to use the new, and unproven products, provided all patients enroll in a registry. The patient’s (de-identified) clinical information (pathology reports and biomarkers, for example) would be included in the registry. This would allow the FDA and researchers to determine, by the end of a pre-set conditional period, whether or not the drug is safe and effective when used on a larger population.
Sounds like a great idea, doesn’t it? It eliminates Phase III efficacy trials, which are slow to complete, expensive to conduct, and drive up drug prices. Musella and Tenenbaum’s plan would give patients ready access to potentially life-saving treatments, speed up approval, reduce the burden on the FDA, bring down the cost of drug development, and create a registry for cancer patients that would open up research avenues. What’s more, Japan, Canada, and South Korea have conditional pathways for new cancer treatments. Why not us?
Because it’s a bad idea that will waste a lot of money and hurt patients. Have we all forgotten the recent history of ineffective and harmful cancer treatments? Autologous bone marrow transplant, or ABMT, and Avastin (bevacizumab) are two that come to mind.
ABMT was developed in the 1980s and was used on at least 41,000 women with metastatic breast cancer. Oncologists, transplanters, and the press embraced the treatment enthusiastically on the basis of a single study comparing it to historical controls. It took more than a decade to accumulate enough patients in randomized controlled trials (RCTs), which showed in 1999 that ABMT was worse than standard chemotherapy—it killed about 10 percent of patients. Insurers paid more than $3.4 billion over the course of the decade for a treatment that actively harmed patients.
A decade after that, Avastin was approved for breast cancer on a fast track in 2008, based on “time to progression.” While there wasn’t any proof that time that progression directly affects how long patients live or their quality of life, Genentech, the manufacturer, argued that it was a good marker of improved patient outcomes. By 2010, worldwide sales of Avastin had hit $6.8 billion.
But once again, it took an RCT to show that the drug caused significant side effects and offered no survival benefit. A few patients may actually have been helped by the drug, but there is no way to predict ahead of time whether patients would be helped or harmed. The FDA withdrew Avastin’s approval for breast cancer treatment in 2011.
Imagine Avastin under a conditional approval plan. Women on the drug would have enrolled in a registry, and that registry might have turned up the side effects that emerged in the actual post-marketing trial. But it could not have shown lack of efficacy.
There are several reasons for this, the main one being that outcomes for people on a registry have to be compared to something. That something would be historical controls–similar patients who have had other treatments. The benefits of most cancer treatments are modest at best, and it’s very easy to be fooled by historical controls unless a new drug has extraordinary power to extend life or cure the disease.
There are other ways to solve the problems of high drug prices and slow development of truly innovative drugs for rare cancers, which Musella and Tenenbaum cite as their motivation for recommending conditional approval for cancer drugs. Congress could let Medicare negotiate drug prices. It could increase the FDA’s budget to speed up approvals. (FYI, the FDA already approves drugs faster than any regulator in the world.) Insurance companies could be required to cover treatment for patients in randomized controlled trials.
Even though Musella and Tenebaum’s registry is a bad replacement for RCTs, it’s still a great idea. Manufacturers can track defects in cars and toasters better than the US tracks outcomes and safety of new medical products. Registries should be required for every patient who receives an implantable device or a new drug for any condition. Let’s use registries for what they’re good at, which is spotting harms once a product is used in a large population. Don’t erode the approval process for the sake of speedy access to drugs of uncertain benefit.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

In Memorium: Richard E. Horowitz, M.D.

by Dr. George Lundberg

Searching for Truth in Cancer Clinical Trials

Richard E. Horowitz, MD, Clinical Professor of Pathology, Keck School of Medicine at USC; Emeritus Professor of Pathology, UCLA School of Medicine; Consulting Pathologist, Los Angeles Veteran’s Affairs Medical Center

Q: Who has been among the strongest supporters and most constructive critics of CollabRx (since 2010) and Curious Dr. George (since 2016)?
A: American pathology lost one of its greatest leaders (and I lost one of my best friends) when Richard Horowitz died on March 15, 2017 in Los Angeles, California. Still of sharp mind and keen humor, he died with dignity and grace, in a manner of his own choice during home hospice care, of metastatic non-small-cell adenocarcinoma of the lung.
Richard and I met across an autopsy table at the old LA County General Hospital in summer 1967. We bonded and remained colleagues and friends who shared many professional beliefs based on personal experiences for 50 years.
Richard was born in Vienna, Austria on May 17, 1931. He left Austria with his parents to flee Hitler’s scourge in 1939.
His college was UCLA, medical school UCSF, internship at Los Angeles County General Hospital, and pathology training (after 2 years in the US Army at Walter Reed doing germfree research) at Mount Sinai in New York. Richard was one of those rare individuals who could keep one foot in academia and one foot in private practice in perpetuity and perform brilliantly in both.
Richard’s “real world” pathology practice management writings and teachings exerted wide influence. But his “tour de force” was always the irreplaceable medical learning uniquely gleaned from his legendary “Organ Recitals” at USC, UCLA, and especially the Wadsworth Veterans Administration Hospital.
Richard achieved many positions in organized pathology, wrote numerous articles, chapters and books, and received many awards.
Family life was very important to Richard; he leaves his lovely and dedicated wife Nona, 3 daughters, and 4 grandchildren.
Richard’s total course of illness after initial diagnosis (malignant pleural effusion discovered at a routine annual medical checkup with established widespread metastases) was 9 months. He tried “Precision Oncology”; his cancer was found to harbor an EGFR mutation, so he was begun on erlotinib. He experienced adverse effects of such severity that he decided to decline further “curative” therapy of any sort and quickly moved into palliative home-hospice care.
Richard’s last publication in a primary source medical journal was in February 2017 in the Archives of Pathology and Laboratory Medicine. Co-authored with Dr. Sarah Bean, it was entitled “Pathology’s Stepchild”, all about the dilemma of Clinical Chemistry and what to do about it.
His final (of many) contributions to the CollabRx discussion group posted on February 25, 2017. It reads:
Sirs & Madams:
You are making pronouncements and decisions based on insufficient knowledge.  Until the use of autopsies becomes the standard of whether the new therapy worked or how the new therapy’s side effects caused the death, we do not have adequate data.  I previously sent the following:

  1. A letter sent to the Wall Street Journal (Published on Sep 23, 2016)
    . . . The autopsy is credible outcome measure; nothing else can attest as convincingly to the accuracy of a diagnosis or the efficacy of a therapy.  Few, if any, clinical trials utilize the autopsy to test their hypotheses . . . .
  2. A short composite of the many autopsies I have personally done:
    The patient has Stage IV lung cancer; all standard therapy has failed.  The patient is coerced into treatment, first with targeted therapy and later, with immunotherapy.  Soon he experienced diarrhea – the oncologist “handles” that with loperamide which results in annoying constipation. Then after a few days, there is marked increase in dyspnea – is it a progression of the disease, perhaps carcinomatous pneumonia or therapy related (auto-immune) interstitial pneumonitis? Well, that can certainly be treated with steroids.  Oh, the oncologist forgot to tell the patient that he needs CNS radiation because of brain metastases.  So the patient is given a course of radiation therapy – unfortunately, there is significant cerebral edema.  Again the oncologist ameliorates that with steroids, however, the cognitive impairment and confusion persist.  About the same time there are some cardiac arrhythmias – are they due to metastases to the heart or due to “auto-immune” myocarditis?  No worry, add some more steroids.   Regrettably a mixed bacterial and fungal pneumonia develops and that, of course, is treated with powerful antibiotics.  Within a brief period of time another bout of diarrhea, this time due to C.difficile, develops and progresses into a dire megacolon that appears about to perforate.  The patient is taken to surgery, the colon has, in fact, perforated and a segment is resected.  In the surgical ICU the early signs of sepsis appear, soon septic shock ensues and the patient dies after prolonged intensive, but futile, care.  The surgeon requests an autopsy, the oncologist does not attend the autopsy and does not answer the call when informed of the Cause of Death.

In September 2015 the National Academy of Sciences/IOM released their Report “Improving Diagnosis in Health Care”. The report listed eight Goals and multiple recommendations. Goal 4 was to develop and deploy approaches to identify, learn from, and reduce diagnostic errors and near misses in clinical practice and Recommendation 4C was that HHS should provide funding…to conduct routine postmortem examinations on a representative sample of patient deaths. It is incumbent on oncologists to obtain autopsies – then they will know if their “magic bullet” worked or killed.
R.E. Horowitz, MD
George Lundberg, MD
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

Fibrolamellar Hepatocellular Carcinoma: Still Rare but Deadly

John R. Craig, MD PhD, Retired pathologist and formerly Medical Director, St. Jude Cancer Center, Fullerton, CA; Member, Board of Directors, FibroFoundation

Q: You were the lead author in a 1980 paper in Cancer that clearly delineated an unusual form of Hepatocellular Carcinoma that you termed “Fibrolamellar Carcinoma”. Now, 37 years later, what insights of importance can you share about this unique malignancy?

A: CRISPR/Cas 9 technology, fruit flies, mice and zebrafish are among the tools being used in numerous academic laboratories, encouraged by the Fibrolamellar Foundation, to determine whether the 400kb deletion found on Chromosome 19 in 90% of patients with Fibrolamellar Hepatocellular Carcinoma (S. Simon PhD, Rockefeller University) is a driver mutation.
In 1980, with renowned liver pathologist co-authors Hugh Edmondson and Robert Peters, I compiled and published the first large series of Fibrolamellar Hepatocellular Carcinoma (FL-HCC) cases in the journal Cancer.
This rare cancer has an annual detection rate of approximately 100-200 patients in the USA and occurs primarily in young adults 15-30 years of age.
After our publication, we received many consultations by pathologists who were eager to share patient information and observation. Over the next 25 years, additional publications introduced unusual findings such as increased serum vitamin B12 binding globulin and other tumor markers, such as des-carboxy prothrombin, and plasma neurotensin. Unfortunately, none of these observations advanced either the detection of the tumor or improved treatment.
Some patients are cared for at academic medical centers, but neither chemotherapy nor radiation treatment has been found to be useful. Complete surgical resection offers the best hope but is usually performed late in the course of the disease since the young patients are often thought to be in good health and have few symptoms.
In recent years, these young patients often connect by social media and have developed Facebook pages. They communicate about their disease, their suffering, treatment options, and acceptance of their disease. There is an annual fall meeting of patients and families to share their experiences.
The family of one young patient (Tucker Davis) answered the plea of their son, and in his honor, in 2008, established the Fibrolamellar Foundation with the goal of finding a cure.
The mutation described above is the result of a fusion of the first exon of DNAJB1 with exons 2-10 of PRKACA. This mutation results in a functional chimeric protein, DNAJ-PKAc, which is highly expressed in almost all FL-HCCs. Little is understood about how the mutant PKA kinase may drive cancer formation.
Numerous academic laboratories are developing models to study this genetic deletion and attempt to learn how it changes the hepatocyte and promotes metastasis. There is hope that treatment may be discovered by interrupting this mutation effect within the malignant cells.
Protein kinases are involved in complex intracellular signaling involving cell proliferation, motility, angiogenesis, anti-tumor immune reactions and other functions. There are more than 518 kinases known within the human genome but the functions of most are not understood. However, small molecule kinase inhibitors are already active in current cancer treatment for chronic myelogenous leukemia, acute lymphoblastic leukemia, and several other cancers of lung and breast. However, no kinase inhibitors are known for this mutant kinase in FL-HCC.
In our initial article, we suggested a possible etiology due to modern industrial life with pesticides or chemicals. But a recent search of old records in a large reference academic center identified some patients with this cancer long before 1940. Thus, our modern industrial contamination may not be a reason for tumorigenesis.
Similar to many other organizations representing rare diseases, the Fibrolamellar Foundation is a philanthropy that has encouraged collaboration by major academic centers and scientific research meetings bringing together diverse scientists to discuss the models and consider investigations. Ultimately, collaborative clinical trials will be necessitated since this malignancy is rare.
We believe that collaborative research with multiple experts in diverse fields who share data and concepts will be necessary in order to apply the knowledge and develop the understanding of how to connect this chimeric protein mutation and ultimately produce an effective treatment.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

A Proposed New FDA Drug Approval Pathway: “Conditional”

 

Al Musella, DPM, President, Musella Foundation For Brain Tumor Research & Information, Inc., Hewlett, NY. Marty Tenenbaum, PhD, Founder and Chair, Cancer Commons, Los Altos, CA

Q: The delay time from discovery/observation, through validation to approval and distribution/use of new cancer treatments remains excessive. With promising experimental treatments, advanced computer technology and biostatistics, creative alternatives to traditional randomized clinical trials, and a government seeking efficiencies, might it now be time for the FDA to issue: “Conditional Approvals”?
A: The first advances in oncology occurred at a time when there were no regulations. Doctors had ideas, and put them to work immediately. They adjusted and combined treatments as needed until they were optimized and became standard treatments. Many types of cancer were cured by this work.
Unfortunately, for patients, with glioblastoma, pancreatic cancer and other rarer cancers, the prognosis remains dire: average survival with currently approved treatments is less than 2 years. These patients can’t afford to wait a decade or more for new drugs to be approved.
The good news is that the oncology drug development pipeline is full of promising targeted- and immuno-therapies that have already demonstrated safety and at least some evidence of effectiveness. However, under current regulations, it will take years before the average patient can get access to these potentially life-saving treatments. Moreover, it is likely that a cure will involve intelligent combinations of treatments. Under current regulations, combination testing cannot even begin until these drugs are approved. And what if a new treatment was not effective as a monotherapy, but could be an essential component of a multi-drug cocktail, say to block a resistance pathway. Catch 22. Under current regulations, many good ideas will never get to patients, and those that do get approved have to be priced so high that many patients cannot afford them.
We would like to propose a new pathway to FDA approval – the “Conditional Approval” – that addresses these issues. It would allow the FDA to approve a treatment that shows safety and a biological effect in a small group of patients. (Click HERE for more details). The twist is that it would require patients using these drugs to participate in a registry where their doctors submit details on the treatments they use, side effects and outcomes.
Conditional approval would be granted to treatments that have been proven safe in a clinical trial(s) with at least 25 patients, and have demonstrated biologic activity: an improvement in a biomarker, brain scan, progression free survival or overall survival.
Once approved, the treatment could be offered as if it had a standard approval, and could not be denied by insurance as being “experimental”. However, all patients who use a conditional treatment would be required to participate in a registry for the duration of the conditional approval period, and to sign a consent form acknowledging and agreeing to the risks inherent in undergoing a treatment whose safety and efficacy have not been fully tested.
The FDA would conduct periodic reviews of this registry data, with three possible determinations: 1) If the safety is questionable or if the results look worse than the standard treatments, conditional approval would be withdrawn, and the manufacturer could continue on the standard paths of approval. However, the FDA could not use these results against the standard approval tracks, as the patient population was not controlled and patients were combining other treatments with it; 2) If the results look at least 20% better than the standard treatments, in the first 50 patients over a predetermined period of time, full approval is granted; or 3) If the results are similar to standard treatments, the conditional approval is maintained until the review shows either the treatment is good enough for full approval or bad enough to withdraw approval.
The decision to try a conditionally approved drug, alone or in combination, would be up to treating physicians, who could consult with peers through a network linked to the registry or use a decision support app that exploits the registry as a database (e.g., show me all treatments and combinations that have been tried on similar patients, sorted by most effective, most cost effective, best risk / benefit ratio, cost, or least side effects.) Such apps could also support low cost point-of-care ‘registry trials,’ whereby patients are dynamically assigned to treatment arms based on expert recommendations and clinical outcomes for similar patients.
It is painfully obvious that the way to cure our currently incurable cancers is to use a combinational approach. We may well have the necessary tools available today—but we are not allowed to use them. When faced with certain death, we believe it is acceptable to not have 100% proven safety and efficacy. We will be approaching the FDA with a request to pilot conditional approval in brain cancer – because life and death decisions should not be made based on regulations – they should be based on what is best for the patient, as determined by the patient and his/her doctors.
We have been working on this plan for a while, but we think now is the time for it to actually be approved. Everything is coming together – like the perfect storm:

  1. We finally have a few experimental treatments in the pipeline that look really good.
  2. The new President is slashing regulations and calling for faster FDA approvals and for slashing drug prices.
  3. Computer technology and biostatistics have reached the point where our plan for a registry trial can be just as reliable as traditional phase 3 trials – maybe more so.

IF this proposal is put into effect, it could lead to rapid advances in the treatment of brain tumors, and the possibility of a breakthrough cure in a few years, instead of the decades it would take on the current path.
We need your support and will be reaching out in a few weeks for help with writing letters and making phone calls. Meanwhile, we welcome your thoughts.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
 

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

AACR: Advances in Immunotherapy to Continue

Srivani Ravoori, PhD, Associate Director, Science Communications; American Association for Cancer Research

Intro: The American Association for Cancer Research (AACR) publishes a forecast blog post at the start of each year to ask prominent cancer research leaders what they envision the new developments will be in areas like immunotherapy, precision medicine, cancer prevention, and health disparities.

In this excerpt from the 2017 post in Cancer Research Catalyst, we interviewed immunotherapy expert Elizabeth Jaffee, MD, on her views on what might develop in immunotherapy this year. Dr. Jaffee is the Dana and Albert “Cubby” Broccoli Professor of Oncology and Professor of Pathology at Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins. She is also the Associate Director of the Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins. Dr. Jaffee was recently named the President-Elect of AACR for 2017-2018.

Q: The American Association for Cancer Research (AACR) is arguably the World’s most important professional organization of volunteers in the cancer field. As we enter 2017, what does AACR consider the field’s greatest challenges and opportunities?

A: “The good news in the field of immunotherapy is that we are learning a lot more about the signals that tumors send to inhibit an effective immune response against them,” says Jaffee, who is a past board member of the AACR.We have already turned this knowledge into therapeutics that inhibit some of these signals (checkpoint inhibitors) so the T cells can be effective in attacking the cancer cells, and developing therapeutics that can activate certain other cells within the tumor microenvironment (checkpoint agonists) to help further activate the T cells, she says.
With these approaches, we have been able to convert some metastatic cancer patients with weeks to live into those with chronic disease living a better quality of life, Jaffee adds. In 2017, we are going to see checkpoint inhibitors being approved for more cancer types and as first-line treatment for some cancers, she notes.
The bad news, however, is that these drugs only work for about 20 to 25 percent of all cancers. Further, these drugs can unleash autoimmunity in patients who respond. The side effects can be controlled currently with steroids in some patients, but this year we will learn more about ways to deliver these drugs in a more targeted way to circumvent the toxic side effects, Jaffee says.
“In 2017, I expect to see the development of new drugs that target additional immune checkpoints,” says Jaffee. One reason why almost 70 percent of cancers do not respond to checkpoint inhibitors is that the cancer cells inhibit different pathways that affect T-cell function. Therapeutics targeting immune-evasion mechanisms other than the PD-1/PD-L1 checkpoint, such as IDO, CD40, OX40, TIM-3, LAG-3, and KIR, are already in early clinical development. We will see them progress to clinical testing, alone or in combination with PD-1/PD-L1 drugs, and some of them may be approved or may come close to approval this year, Jaffee predicts.
This year, we will also see a lot of preliminary data identifying new biomarkers of immunotherapy response, according to Jaffee.

AACR Immunotherapy 2017 What Advances Can We Expect

 
Other approaches to get more patients to respond to immunotherapies include activating T cells using vaccines, radiation therapy, or different types of immune-activating chemotherapies, Jaffee says. Combining immune checkpoint inhibitors with agents that can help uncover cancer antigens, such as PARP inhibitors that can make new tumor antigens available to the T cells, or epigenetic agents that can turn on the expression of certain proteins, is another avenue. “We will start seeing results from such studies this year,” Jaffee notes.
We are likely to make more progress this year in personalizing cancer treatment with vaccines, Jaffee predicts. “We are starting to understand the importance of neoantigens for targeting by the immune system,” Jaffee notes. Tumors of many patients who respond to immunotherapy create neoantigens constantly. If we can identify them by sequencing the tumors, we can develop vaccines against them to jump-start the immune system, she says. “We are going to see several clinical trials trying this approach this year.”
“As a member of the Blue Ribbon Panel, one of the 10 areas we identified as at the point of making huge progress is basic research to better understand the mechanisms behind immunotherapy response,” says Jaffee. Answers to questions such as, “What makes a pancreatic cancer that doesn’t respond to immunotherapy different from melanoma that responds to immunotherapy?” or “Why do some tumors that have the biomarker of response not respond while some that do not have the biomarker respond?” or “How to make CAR T-cell therapy work in solid tumors?” can only be found by pursuing more basic science research, she notes.
“We have the technology to find answers to many basic research questions and there is excitement among academia, industry, and federal agencies to work together; however, we need more funding to pursue such important studies,” says Jaffee. While she is concerned about the uncertainty regarding the scientific priorities of the new administration, she is cautiously optimistic.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Curious Dr. George | Plumbing the Core and Nibbling at the Margins of Cancer

A Fully Integrated Histo-Molecular Pathology Report

Margaret L. Gulley, MD, Professor of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill

Q: The CAP, ASCO, ASCP and AMP have developed guidelines for interpretation and reporting of NGS variants in cancers. These analyses are performed in a wide range of types of labs and involve professionals from many disciplines. The reports should take into consideration the bioinformatics, molecular data, source of specimen, age, gender, and location of patient, treatment background, morphologic diagnosis, immunohistochemical tumor profile, and special stains. Who is the best qualified and positioned person to consolidate all of this information and produce a final integrated pathology report and how should this be accomplished?

A: Pathologists are the best professionals to synthesize data from all the tests done on a given tumor specimen via an integrated report that is actionable for downstream medical decision-making.
Hematopathologists enthusiastically embrace new technologies and they are the role models for data integration and analysis of microscopy, flow cytometry, histochemistry (IHC, ISH, FISH), karyotype, PCR, or sequencing.
College of American Pathologists guidance for reporting molecular test results suggests that all results on a given tissue be synthesized by one pathologist, typically the histopathologist although increasingly the molecular pathologist who performs ancillary genomic testing. Since molecular results are best interpreted in the context of histomorphology of input tissue (e.g. percent malignant cells), and in the context of the clinical dilemma to be solved by the test, good communication is essential to assuring that professionals selecting the tissue block, doing the test, and interpreting results provide answers to pertinent medical questions. Access to patient medical records promotes high quality interpretation of histo-molecular findings.
Pathologists are in the best position to allocate precious (often small) specimens and to prioritize which ancillary tests are most critical in a given clinical scenario. Certain tests are feasible only on selected specimen preparations (e.g. karyotype requires fresh tissue), and the pathologist is vital to assuring that tissue is processed in a manner that maximizes success.
In many cases, ancillary tests are ordered by the surgical pathologist who feels comfortable synthesizing results of the test with histomorphology, even if different professionals (e.g. cytogeneticists, molecular pathologists) performed and interpreted raw data. What about ancillary tests ordered by clinicians, sometimes months or years after the microscopist issued their histopathologic interpretation? Specimen requirements must be considered (e.g. fixative type, size, % malignant cells) along with whether to test primary vs metastasis, recent biopsy vs an older or larger resection, in situ vs invasive components, etc. There needs to be better compensation for the expert work of the surgical pathologist to understand the clinician request in order to retrieve and select the best archival tissue portion for the test. Compensation is also needed for the resources required to incorporate test findings into a revised integrated report.
Ancillary tests have benefits and limitations that are best understood by the testing laboratory, so it may be realistic for the testing laboratory to synthesize data from the surgical pathology report, rather than expecting the surgical pathologist to integrate lab data. When a histopathologist feels uncomfortable performing an integrative interpretation of lab data, the histopathologist should at least make it clear in their (addendum) pathology report what test(s) were ordered on which block in order to facilitate work by another professional in synthesizing findings and minimizing accidental repeat testing.
The criteria pathologists use for tumor diagnosis and classification are evidence-based and are updated periodically by various professional groups. Increasingly, ancillary tests are value-added components of standard-of-care surgical pathology workups. Since every patient is different and every tumor is different, pathologists should be valued for their expert judgment and for taking responsibility for translating histo-chemical findings into expert consultations.
Copyright: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.