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

How an Expert Would Manage His Own Advanced Bladder Cancer: An Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Genitourinary Oncologist and Researcher, Beth Israel Deaconess Medical Center; Instructor in Medicine, Harvard Medical School

When facing a frightening new cancer diagnosis, some people ask their doctors, “What would you do if you were me?” Here, our Curious Dr. George asks Daniel E.C. Fein, MD, how he would handle his own case of advanced bladder cancer. Dr. Fein is a genitourinary oncologist at Beth Israel Deaconess Medical Center (BIDMC) in Boston, MA, as well as an Instructor in Medicine at Harvard Medical School. (Note: This piece originally appeared in May 2023; now, Dr. Fein provides his updated expert perspective, with new information on preferred treatments for people with bladder cancer.)

Curious Dr. George: Imagine a hypothetical scenario in which, as a non-smoking, very busy genitourinary medical oncologist, you were surprised to experience painless gross hematuria. Rapid subsequent microscopic urinalysis confirmed many red blood cells and some white blood cells. Urine cytology was suspicious for malignant cells, not otherwise specified. A complete blood count and blood chemistry panel were normal. At cystoscopy a sessile mass was found and biopsied, revealing a high-grade urothelial carcinoma. You then recognized a 10-pound loss of weight. The upper urinary tract was normal by imaging studies, but your pelvic lymph nodes were enlarged, and three suspicious lesions were found in your left lung. How would you proceed?

Daniel E.C. Fein, MD: Although I care for many patients with bladder cancer and have been intimately involved in the medical field for some time, I can only imagine how I might react to such shocking news. Why me? What did I do wrong? How should I tell my family? Do I need to take time off work? My mind would be spinning out of control.

I would try to take a deep breath and tell myself that it is likely that I have been diagnosed with an advanced bladder cancer. I would know that although this is likely incurable, there are new treatments available that can help me live longer and improve my quality of life. While establishing care with a cancer treatment team, I would reach out to my primary care doctor and therapist to disclose to them this new diagnosis and ask for the best way for me to manage my mental and physical health while beginning my journey.

Some background: Bladder cancer accounts for approximately 4% of new cancer cases in the US, with around 10% of those presenting with metastatic disease. Most cases are confined to the bladder and can be treated with resections and medicines instilled into the bladder. When cancer invades into the muscle of the bladder wall, patients can be potentially cured using a combination of treatments, such as chemotherapy followed by removal of the bladder, or chemotherapy with radiation. When bladder cancer has spread to distant sites outside of the area of the bladder (such as the lung) they are typically not curable but can be controlled with a combination of medications to help patients live in the range of years.

Before treatment: The approach to treating advanced bladder cancer is changing rapidly, with new approvals of medications and combinations coming out every year. Because of this, I would seek care at a nearby academic center that has experience treating bladder cancer using a multidisciplinary team of urologists, medical oncologists, and radiation oncologists.

I would undergo a biopsy of a lung lesion to confirm this is metastatic bladder cancer. I would direct this biopsy to be sent for molecular testing for gene mutations in FGFR2/3 and for expression of the marker HER2, which would tell me what kind of treatments I could receive in the future. I would also undergo germline genetic testing to see if I was born with a gene mutation that made me more likely to get this cancer and potentially others. I would also ask if I should have any additional imaging, such as an MRI head, as this may be helpful for some patients.

Choosing treatment: Because of how quickly this field is changing, I would ask my care team if there were any clinical trials in my region with new and promising treatment combinations available. As a member of the Innovation in Cancer Program at BIDMC, I have seen firsthand the outstanding care provided to cancer patients on clinical trials and would certainly participate in one if it was right for my cancer treatment and quality of life.

If I thought a clinical trial wasn’t right for me, I would ask if I could be a candidate for the newly FDA-approved combination of enfortumab vedotin and pembrolizumab (EV + P), a combination of a targeted chemotherapy and immunotherapy. Because of a recently published clinical trial, this combination is now considered the preferred first-line therapy for all patients with metastatic bladder cancer.

If I was unable receive EV + P because of other medical conditions (such as severe autoimmune disease, difficult to control diabetes, or severe peripheral neuropathy), I would ask if I could receive a platinum-based chemotherapy combination. Conventional platinum-based chemotherapy (cisplatin or carboplatin with gemcitabine, or dose-dense MVAC) has been the standard-of-care treatment for advanced bladder cancer for many years and can still be an option for some patients if they are unable to receive or decline the EV + P combination. In recent years we would give at least four cycles of platinum-based chemotherapy, and if there is some control of the cancer, switch to a maintenance immunotherapy called avelumab. There is actually another combination of platinum-based chemotherapy with an immunotherapy called nivolumab that was also recently approved based on a clinical trial, but because of how effective enfortumab vedotin plus pembrolizumab seems to be, I generally recommend EV + P first.

What next?: I may have challenges with managing side effects of treatment, coping with my new diagnosis and limited life expectancy, or sharing my experiences with friends and family members. I would seek assistance from a palliative care team and a local cancer support group who can focus on these mental, emotional, and physical challenges to improve my quality of life.

A new diagnosis of cancer, at any stage, can be life changing. If you are going through this, please know that you are not alone and that there are people around the world with and without cancer who have dedicated their lives to making yours better.

Dr. Fein can be reached at dfein@bidmc.harvard.edu or on Twitter at @DFein_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

How an Expert Would Manage His Wife’s Metastatic Breast Cancer: An Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Senior Medical Director, Breast Morpheus/Giredestrant, Genentech Product Development Oncology

For most people with breast cancer, a lump is the first sign that leads to diagnosis. But some cases are not detected until after spread has already occurred. Here, our Curious Dr. George asks Richard B. Schwab, MD, how he would approach such a case if his own wife were the patient. Dr. Schwab is Senior Medical Director at Genentech Product Development Oncology. (Note: This piece was first published in October 2021. We now present it with Dr. Schwab’s updated affiliation. His reply below remains up to date and relevant for people dealing with advanced breast cancer.)

Curious Dr. George: How would you, as a breast cancer expert, manage your wife’s cancer if it were to present as shortness of breath or back pain, with a breast mass only then detected?

Richard B. Schwab, MD: Doctors frequently take care of their family members for minor problems; cancer is not one of them. Even caring for patients, managing one’s own anxiety about the normal uncertainties of medicine is not easy. Managing of my wife’s cancer would not be possible, so I would have one of my excellent colleagues be her oncologist.

That said, I would be answering my wife’s questions about this frightening situation and trying to guide her. She gets her screening mammogram every year, so presenting with metastatic breast cancer would be very unusual. Only 5% of breast cancer is de novo metastatic, and many of these patients did not have recommended screening before diagnosis. However, screening mammograms are not perfect, so your hypothetical scenario is possible.

If she was short of breath, I think we would utilize the emergency department. Although cancer is usually a relatively slow process, there are rare times when patients can become very ill during the normal time required for diagnosis and treatment initiation. CT scans can be done quickly and could identify a pulmonary embolism, pleural effusion, or lymphangitic spread. Starting appropriate therapy quickly (anti-coagulation, thoracentesis, or chemotherapy respectively) could be critically important.

Making a pathologic diagnosis would be the next step, and would radically alter treatment (and life) plans. Breast cancer is not one disease. Oddly we would be hoping for an aggressive estrogen receptor-negative and HER2-positive cancer. These cancers are highly responsive to numerous treatments, and cures—even with metastatic disease—are becoming more and more common. A triple-negative cancer would be complicated. Some patients with triple-negative breast cancer, particularly limited to the lungs, do end up cured, but these cases are rare and overall this type of breast cancer has the fewest treatment options. Last but most common, particularly if her cancer had spread to the bones, would be estrogen receptor-positive disease. These cancers generally grow more slowly and on average patients survive longer with this type of disease. However, these cancers are never truly cured, although for older patients lifelong disease control can sometimes be obtained. For a patient as young as my wife new therapies would be needed to have any reasonable hope of lifelong disease control.

Which location to biopsy is a common challenge we would need to address. Biopsy of the breast is the easiest, but may not reflect the more dangerous disease that has spread to other organs. Lung biopsy has some additional risk, it is anxiety provoking, and sample quantity (or even successful sampling) can be challenging. Biopsy of the bone, while very safe, can give unreliable results due to the need to decalcify the sample prior to testing. The details of the patient’s case and the expertise of the doctor performing the biopsy matter. Having expert trusted colleagues in radiology and pulmonology would be a significant advantage for my wife.

I mentioned the idea of truly curing my wife. I define true cure as a lack of cancer progression for at least 5 years after stopping therapy. I have cared for many patients with metastatic disease that has been cured, and that knowledge would help my family get through what is fortunately only a hypothetical situation.

Dr. Schwab can be reached at schwab.richard@gene.com.

***

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

How an Expert Would Manage Her Own Advanced Colorectal Cancer: An Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Christina Wu, MD
Professor of Medicine, Mayo Clinic, Phoenix

When facing a new cancer diagnosis, some people ask their doctors, “What would you do if you were me?” Here, our Curious Dr. George asks oncologist Christina Wu, MD, how she would handle her own diagnosis of metastatic colorectal cancer. Dr. Wu is a Professor of Medicine at the Mayo Clinic in Phoenix, Arizona. (Note: This piece originally appeared in 2020; now, Dr. Wu provides her updated expert perspective on molecular testing, targeted therapies, surgery, and clinical trials.)

Curious Dr. George: As an experienced academic and practicing clinical oncologist at the Mayo Clinic, you have particular interest, training, and experience in colorectal cancer. What would you do if you personally were discovered to have an asymptomatic, unsuspected, non-obstructing adenocarcinoma of the ascending colon that had already metastasized to your liver?

Christina Wu, MD: Colorectal cancer is one of the most common cancers in the U.S., and there has been a noticeable rise in early-onset colorectal cancer. Every patient with metastatic colon cancer to the liver is uniquely assessed because there are systemic and liver-directed treatment options. If I were diagnosed with colon cancer and liver metastasis, I would want a multi-pronged approach to include the following:

Next-generation sequencing (NGS): First of all, I would want both blood and tumor NGS testing. Blood NGS results can be back within a week, and tumor NGS is more expansive and complete. The tumor biomarkers I would want to know first are mismatch repair (MMR) protein or microsatellite instability (MSI) status. If deficient MMR or MSI-high, I could benefit from immunotherapy (pembrolizumab or nivolumab with or without ipilumumab) and be spared chemotherapy. Other tumor markers include KRAS/NRAS/BRAF mutation (mt), HER2 amplification, and NTRK and RET gene fusion.

I would be eligible for anti-EGFR antibody (panitumumab or cetuximab) in combination with doublet chemotherapy (FOLFOX or FOLFIRI) if my tumors were KRAS/NRAS/BRAF wild-type (wt) and HER2-negative with a left-sided primary cancer. HER2-targeted therapy such as tucatinib and trastuzumab could be a chemotherapy-free option if the tumor was KRAS/NRAS/BRAF wt and HER2 positive. Trastuzumab-deruxtecan could also be offered if I was HER2 positive. A BRAF V600E mt would be targeted with encorafenib and an anti-EGFR antibody. A KRAS G12C mt would be targeted with sotorasib/adagrasib and cetuximab/panitumumab. If I had the rare NTRK fusion mutation, I could be treated with entrectinib, larotrectinib, or repotrectinib. RET gene fusion could be targeted with selpercatinib.

Multi-disciplinary tumor board: I would want high-quality imaging and experts from medical oncology, surgical oncology, colorectal surgery, radiation oncology, gastroenterology, radiology, and interventional radiology reviewing my case in a tumor board. It would be meaningful to know upfront whether the liver metastasis was surgically resectable, because I may opt to receive systemic chemotherapy followed by surgical resection. If I required conversion therapy or reduction in my tumor burden, I would consider triplet chemotherapy (FOLFOXIRI) or systemic targeted therapy to get more of a response so that I could get to surgery. However, if I had clearly unresectable disease, I would choose systemic targeted therapy or doublet chemotherapy, such as FOLFOX or FOLFIRI and bevacizumab, to improve my quality of life.

With unresectable disease, I could also consider radiation, Y90 treatment, or ablation to the liver tumors, if I had good control or surgical resection of the extrahepatic disease. In addition, liver transplant may be a future consideration if I had well-controlled, unresectable liver metastasis, with no evidence of extrahepatic metastasis. Although not present at a tumor board, the palliative care team would be a crucial component of my care.

Genetic counseling: Some hereditary syndromes lead to colon cancer development, including Lynch syndrome, familial adenomatous polyposis, Peutz-Jeghers syndrome, and juvenile polyposis. I would choose to pursue germline testing to screen for such conditions.

Clinical trials: There are studies looking at targeting KRAS mutations, bringing targeted therapy to the first-line setting, and providing immunotherapy combinations. I would definitely want to expand my therapy options by enrolling in clinical studies.

Final thoughts: Systemic and liver-directed treatment options are evolving, and thus a multi-disciplinary approach would be essential from the moment of my diagnosis.

Dr. Wu can be reached at wu.christina@mayo.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.

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

How an Expert Would Treat His Own Advanced Lung Cancer: An Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Howard (Jack) West, MD, FASCO
Vice President, Clinical Development, Summit Therapeutics

A new cancer diagnosis is overwhelming. Patients often ask their doctors, “What would you do if you were me?” Here, our Curious Dr. George asks lung cancer expert Howard (Jack) West, MD, FASCO, how he would handle his own diagnosis of advanced lung cancer. Dr. West is a Princeton- and Harvard-educated oncologist with additional training and experience in Boston and Seattle focusing on lung cancer. He is now Vice President of Clinical Development at Summit Therapeutics. (Note: This piece was first published in March 2021. We now present it anew with Dr. West’s updated affiliation. His reply below remains up to date and relevant for people dealing with advanced lung cancer.)

Curious Dr. George: What would you do if you personally were discovered on a routine chest X-ray to have a unilateral pleural effusion which was found by cytopathology to contain mixed squamous and adenocarcinoma cells? How would you proceed?

Jack West, MD, FASCO: Though I’m a never-smoker, we know that is no guarantee of immunity from lung cancer, the cancer type I’ve focused on for the past two decades. Perhaps I develop a persistent cough and worsening shortness of breath over a few months. I get a chest X-ray that shows a large right pleural effusion, and a same-day chest CT confirms this and bilateral lung nodules, perhaps along with several enlarged right hilar and mediastinal lymph nodes. The effusion is drained, and the pathologist gives my doctor and me the immediate feedback that this is a carcinoma, and we quickly learn that the immunohistochemistry profile is consistent with an adenocarcinoma. Where do we go from here?

My next step is to order broad next-generation sequencing, which may entail a new biopsy, either CT-guided or an endobronchial ultrasound and biopsy of whatever is accessible. We need sufficient tissue to send off for broad molecular testing that includes a look for all of the growing collection of lung cancer “driver mutations.” These include long-established markers like a mutation in the epidermal growth factor receptor (EGFR) gene or a rearrangement in the anaplastic lymphoma kinase (ALK) gene, but now also a rearrangement in ROS1, mutation of BRAF V600E, a MET exon 14 skipping mutation, RET fusion, or fusion in the TRK gene. These genetic alterations are found in approximately 0.5% to 10% each in patients with non-small cell lung cancer (NSCLC) and far more commonly in patients with a non-squamous NSCLC tumor. They all have FDA-approved oral targeted therapies with efficacy that generally exceeds what we could expect with our best standard non-targeted approaches involving immunotherapy with or without chemotherapy. Most of these targeted therapies also better tolerated and can work for a prolonged period that may reach the range of years.

This mutation testing typically takes at least 2 to 4 weeks, but the importance of identifying one of these mutations, when present, makes it critical to seek this information at the time of initial diagnosis. Moreover, in addition to the array of markers we currently have targeted therapies for, we expect an FDA approval for an inhibitor of KRAS G12C-mutated NSCLC, seen in about 12% to 13% of advanced NSCLC, in the coming months; all in all, comprehensive molecular testing guides us to an optimal targeted therapy for at least 20% of patients, and that proportion will continue to increase as new targets with effective therapies become available.

In the hypothetical scenario of my own diagnosis, as this testing is being done we’re also testing for tumor PD-L1 expression, which identifies tumors most likely to respond well to immune checkpoint inhibitors, potentially sparing patients first line chemotherapy if they don’t receive a targeted therapy. And I’d seek to complete imaging with a PET/CT and brain MRI, in order to identify whether the cancer has spread to other sites beyond those already identified.

Once these tests are completed, I’d prioritize a targeted therapy if my tumor harbors a driver mutation. If not, I’d generally favor pembrolizumab monotherapy if my cancer is among the approximately 28% to 30% that demonstrates high tumor PD-L1 expression (greater than 50%). Otherwise, if my cancer has neither a driver mutation nor high tumor PD-L1 expression, I’d generally favor a platinum-based chemotherapy doublet with pembrolizumab—an option I would also favor in a patient with a tumor that doesn’t harbor a driver mutation and with high tumor PD-L1 if that patient had many cancer-related symptoms or otherwise showed a pattern of rapid progression in the early weeks of the workup. Though I’d be happy to sequence the immunotherapy with subsequent chemotherapy in patients in whom I’m confident they will still have the performance status to tolerate platinum doublet chemotherapy after progressing on chemotherapy, I’d favor “front-loading” with chemo-immunotherapy together in patients in whom I’m concerned I may only have “one shot on goal.”

Dr. West can be reached at JackWestMD@gmail.com or on Twitter at @JackWestMD.

***

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

How an Expert Would Manage His Own Advanced Prostate Cancer: An Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Marc B. Garnick, MD
Gorman Brothers Professor of Medicine at Harvard Medical School and Beth Israel Deaconess Medical Center

When facing a new cancer diagnosis, some people ask their doctors, “What would you do if you were me?” Here, our Curious Dr. George asks Marc B. Garnick, MD, how he would handle his own advanced prostate cancer. Dr. Garnick is the Gorman Brothers Professor of Medicine at Harvard Medical School (HMS) and Beth Israel Deaconess Medical Center in Boston, MA. He is also Editor in Chief of the HMS Annual Report on Prostate Diseases. (Note: This piece was first published in June 2022 and has now been updated with additional information about neuroendocrine elements, mismatch repair deficiency, and cardiovascular considerations.)

Curious Dr. George: In 2021, you shared with our readers how you, as an expert, would proceed with handling your own severe back pain and spine lesions.

Let’s revisit that hypothetical scenario with some new developments: Further rapid lab studies demonstrated normal peripheral blood counts and no abnormal serum proteins. The back pain stabilized. Digital rectal exam discovered an enlarged, diffusely firm prostate gland with scattered hard foci. Ten transrectal prostate biopsy cores all contained adenocarcinoma with varying degrees of dedifferentiation, Gleason score of 8, repeat PSA was 25.

How should the pathologist handle the remaining tissues? What procedures should a reference laboratory perform? How would genomic, metabolomic, or proteomics inform next steps? And would imaging be helpful?

Marc B. Garnick, MD: With these new developments, the differential diagnosis has now been narrowed. The likelihood of the bone lesions being related to a plasmacytoma or multiple myeloma, though not completely excluded, are less likely. The lead diagnosis now has been established to be a Gleason score 8 prostate adenocarcinoma, which I presume is a Gleason 4+4. Please recognize that based upon the International Expert Consensus on Prostate Cancer nomenclature, this is now best described as Grade Group 4 (GG 4). Grade Group 1 (GG 1) contains Gleason patterns 3+3; GG 2 contains Gleason patterns 3+4; GG 3 contains Gleason patterns 4+3; GG 4 contains Gleason patterns 4+4; and GG 5 contains Gleason patterns 4+5 or 5+5. If this patient did not have suspected metastases to the bones (and even in the setting of metastatic prostate cancer), the pathology biopsy note should mention the presence of any intraductal component, perineural invasion, or lymphovascular invasion, all of which commonly accompany high-grade Gleason cancers. Neuroendocrine elements, although uncommon, can also accompany high-grade Gleason scores. If present, such transformation following treatment to neuroendocrine/small cell-like pathologies represents an ominous development, which is often accompanied by visceral involvement and the need for systemic chemotherapy.

Recent recommendations now suggest that genomic profiling of the patient both for germline and somatic mutations be assessed. The importance of BRCA (either germline or somatic) mutation has increased in significance, especially since there are emerging associations of prostate cancer being found in families of women with BRCA mutations. The finding of a BRCA2 mutation is associated with a more aggressive biology and could help inform treatments at some later day with the PARP class of therapeutics of platinum-containing agents, given the increased sensitivity of BRCA + cancers to respond to these therapies. Likewise, the presence of microsatellite instability (MSI high, or MSI-H) or mismatch repair deficient (dMMR) mutations may prompt treatment with an immunotherapy agent such as pembrolizumab.

The above consideration of more precise pathologic assessment is more important in a patient who has a GG 4 or 5, in whom there is no obvious evidence of metastatic disease. In such cases, one is trying to assess a probability of the patient having subclinical metastatic disease or being likely to develop metastatic disease. For the patient with already-established evidence of abnormal foci (in this case, the bone lesions), we in the past would have assessed with an Axumin scan; now, the availability of either Ga or F linked prostate-specific membrane antigen (PSMA) scanning would be indicated, both for diagnostic purposes as well as assessing down-the-line utility of PSMA-specific therapeutic ligands, such as Lutetium 177.

An assessment of cardiovascular history is increasingly important in helping determine a treatment program that helps minimize any therapy-induced increase in morbidity. Traditional androgen-deprivation therapy (ADT) has long been associated with cardiovascular issues, and now, with the standard addition of second-generation androgen receptor signaling inhibitors to ADT, the cardiovascular risk can double and even quadruple. Patient evaluation by experts in cardiology, internal medicine, or the emerging field of cardio-oncology is becoming increasingly important.

Curious Dr. George: What additional testing and what therapy would you deem to be most appropriate at this point?

Marc B. Garnick, MD: Please recall that this patient (hypothetically me) presented with back pain and multiple bony lytic lesions. Regardless of the etiology of these lesions, identification of any impending spinal cord compression must be assessed. While spinal cord compression is an unusual manifestation of de novo metastatic prostate cancer, it is clinically more common in the setting of treated prostate cancer that has progressed to become castration resistant. I would reassess the anatomic locations of the bone lesions, and then obtain a spinal MRI directed diagnostic evaluation, along with a good neurological examination to inform preemptive radiation or surgical considerations if either impending or actual compression is found.

For treatment of newly diagnosed presumably castration-sensitive metastatic prostate cancer, we now have multiple advances that have been established in this disease setting. First-line ADT (generally with an LHRH agonist or GnRH antagonist) is now supplanted with second-generation agents, such as abiraterone (+ prednisone) or enzalutamide, or other androgen-receptor signaling inhibitor agents, such as apalutamide. ADT + chemotherapy with docetaxel is also appropriate. Given the bone pain and potential neurological issues here, I would select a GnRH antagonist (either degarelix or relugolix) as the preferred therapy.

Dr. Garnick can be reached at mgarnick@bidmc.harvard.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.

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

The Personalized Power of the “N-of-1” Approach

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Glenn Sabin
Director of Strategy & Business Development, Cancer Commons and Author of n of 1

Randomized, controlled clinical trials with a large N—number of participants—are the recognized “gold standard” of evidence-based medicine. Even so, the results of large-N trials can only reveal population averages, and are not predictive of any individual’s response to a given treatment. On the other hand, one can consider every patient with cancer as the sole participant in their own clinical trial with an Nof1, even if the trial is not officially registered, reported, or supported like a traditional trial.

Here, our Curious Dr. George discusses the value of the “n-of-1” approach with Glenn Sabin, director of strategy and business development at Cancer Commons. Sabin is a 33-year cancer thriver and author of n of 1: One Man’s Harvard-Documented Remission of Incurable Cancer Using Only Natural Methods.

Curious Dr. George: Among the many people whose cancer progresses beyond guideline-based standards of curative care, how might one become an n-of-1 trial participant, if they so choose? What actions should be taken by the physicians who bear responsibility for such individuals who desire to further collective knowledge—and their own welfare—by voluntary participation in use of investigational drugs or drug combinations, or off-label use of drugs approved for other contexts by the US Food and Drug administration (FDA)?

In my view, the n-of-1 approach is clinical care and should be paid for by reputable insurance companies. And, the patient can provide fully informed consent, even with the proviso that the process and outcome be shared for educational purposes. Could this approach become a nimble form of adaptive clinical trial? What are its pros and cons?

Glenn Sabin: From my perspective, randomized, controlled trials (RCTs) will continue to be important for population-based investigational cancer drug development—especially when effectively translated to clinical practice.  Standard-of-care treatment that follows guidelines from the National Comprehensive Cancer Network (NCCN) remains effective for many patients who host early-stage and more indolent disease.

However, for those living with advanced and rare cancers—and for whom standard care has not enabled deep and durable remissions after multiple lines of treatment—it is critical to consider a more personalized approach to testing and treatment.

After all, there is only one host (the unique person living with cancer) and one tissue/tumor type (no two malignancies are exactly alike). This is why we are each an Nof1—an experiment of one. This is the distinction between population-based investigations (RCTs) and truly personalized cancer care (Nof1): averages versus individualization.

I see a future close on the horizon where each patient goes through a deep set of diagnostic testing beyond today’s molecular testing (which itself is not used nearly often enough). The resulting data, consisting of multiple datasets, are then precisely interpreted to inform—with the assistance of artificial intelligence—the patient’s best option for their next treatment. Then, the chosen therapeutics are secured, typically in combinations and often as off-label, and administered to the patient.

Alas, in today’s reality, this type of n-of-1 care—even among the most affluent, educated and connected patients—is largely inaccessible. Access is key.

Novel Testing
Testing beyond certain FDA-approved molecular (sequencing of DNA/RNA) and other diagnostics is not currently covered by Medicare or commercial insurance. Those interested in functional testing—the testing of their own tumor tissue across scores of drugs and combinations—need to pay out of pocket, and some of these novel assays can get quite expensive.

Interpretation of Testing Results
These novel tests produce lots of data. Now what? The problem is that very few oncologists can interpret beyond the “targetable” biomarkers and mutations shown in molecular sequencing reports, which typically cover a small set of genes. Add to the mix novel multi-omics assays (looking at proteins, metabolism, the microbiome, and more) and functional testing, and oncologists are not trained to review in a way that supports the treatment decision-making process. Third-party professional services are needed.

N-of-1 Physicians
Oncologists are trained to deliver standard-of-care cancer treatment as defined by NCCN guidelines. Across the US, some oncologists within community practices will consider prescribing outside of these guidelines when patients have relapsed multiple times. Other oncologists, especially within academia, will only depart from NCCN guidelines under an institutional review board (IRB) and investigative new-drug (IND) study setting, even if such a study is created for only one patient (N of 1). In general, oncologists need internal institutional support to prescribe off label, both from their tumor board colleagues and from colleagues who ensure they feel comfortable about potential toxicity when using drugs with limited or no studies capturing potential for severe adverse events.

Drug Access
Today, approximately 30% of all FDA-approved anticancer therapeutics are prescribed off label. Physicians always have the option to prescribe in this way. One major issue is getting off-label drugs covered by Medicare or commercial payors. Otherwise, providers must advocate for their patients (without reimbursement for their efforts) to apply to various programs such as Expanded Access and Compassionate Use. And there is no financial incentive to do so because drugs secured through these channels cannot be marked up.

Data Collection and Registry Studies
The most effective and efficient way to capture n-of-1 managed and treated patient data is within an IRB-compliant registry connected to a learning health system. This approach collects real-world evidence and allows us to learn from each patient in near-real time. The gold-standard comprehensive learning health system does not yet exist, but I understand that progress is moving apace.

Making N-of-1 Cancer Care a Reality
At Cancer Commons, we are actively pursuing solutions to making comprehensive, end-to-end, n-of-1 patient navigation and treatment a reality. Every day we are working on eliminating the obstacles to access, in service of improving the human condition for those hosting advanced disease, and their families and loved ones.

Progress is inexorable.

Mr. Sabin can be reached at glenn.sabin@cancercommons.org.

***

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

How Would an Expert Manage His Own Acute Myelogenous Leukemia: an Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Adam Asch, MD
Professor of Medicine; Nancy Johnson Records Chair of Oncology, University of Oklahoma College of Medicine

When facing a frightening new cancer diagnosis, some people ask their doctors, “What would you do if you were me?” Previously, our Curious Dr. George asked leukemia expert Adam Asch, MD, how he would handle his own hypothetical case of acute myelogenous leukemia (AML). Now, Dr. Asch provides an updated answer, highlighting the new option of clinical trials offering novel targeted therapies for some AML patients. Dr. Asch is Professor of Medicine and Nancy Johnson Records Chair of Oncology at the University of Oklahoma College of Medicine.

Curious Dr. George: Please consider this hypothetical scenario: You are an active clinical oncologist in generally good health, but lately, you have lacked energy, have been feeling a little more tired than usual, have some dizziness on exertion, have been bruising easily, and have lost a bit of weight without dieting. So, you decide to visit your primary care physician for a checkup. On physical examination, she notes that you are a little pale, finds arm bruises, and feels the tip of your spleen. She orders several lab tests. Your hematocrit is 38, hemoglobin 12, white blood cells (WBCs) 49,000 per microliter with many blasts, many promyelocytes, myelocytes, eosinophils, and basophils. Platelets are low. A diagnosis of acute myelogenous leukemia (AML) is made. How would you proceed?

Adam Asch, MD: This would be a fascinating case to discuss in the abstract. But in the first person, as you have been exploring in your Curious Dr. George series, diagnosis and therapy take on a special and sometimes personal urgency. For this 70-year-old oncologist, the hypothetical is a timely one and for the field a relevant one, since AML is, in so many ways, a disease of aging.

For most AML patients, I find that the most important question to ask is if there is a curative intervention. For some, chemotherapy induction and consolidation may lead to a prolonged remission and perhaps cure. But for some disease categories—notoriously, p53-mutated disease—response to traditional chemotherapy is poor. Second, assuming an initial remission follows an induction regimen, how a remission is consolidated is another critical question. For people with high-risk AML (complex cytogenetics, FLT3-mutated) or intermediate-risk AML, allogeneic transplant remains the best means of consolidating a remission and offering a cure to many.

To a great extent, treatment for AML is informed by genetic and molecular studies that have become standard in the field. Cytogenetics to assess chromosomal defects, molecular analysis to identify potentially targetable mutations, and multi-channel flow cytometry to identify a blast phenotype that can be followed to assess measurable residual disease post induction and consolidation are all standard. Translocation 15;17 or “acute promyelocytic leukemia” (APL)—once considered to be the worst type of AML prognostically—is now routinely treated with all-trans retinoic acid and arsenic trioxide, with long-term survival now approaching 90%. The clinical presentation in this hypothetical case does not sound like APL, though the bruising raises the question, and a coagulation profile, if indicative of disseminated intravascular coagulation, might make that more of a question.

Other good prognostic cytogenetics include the translocation of chromosomes 8 and 21—also known as t(8;21)—and inversion of chromosome 16. Each of these has a historical long-term survival rate of about 60 to 70% with standard induction chemotherapy. All other cytogenetic features are classified as intermediate risk or poor risk, with long-term survival rates of 30 to 40% or 10%, respectively. For each of these latter risk groups, transplant remains the only real chance of relapse-free, long-term survival.

Now, before we discuss treatment regimens for AML in general, there are some features of my presentation that would lead me to ask for some additional data. The presence of a spleen tip and the presence of early myeloid forms, eosinophils, and basophils in addition to blasts raise the possibility that my disease is a Ph+ blast crisis rather than de novo AML. If so, treatment with a kinase inhibitor directed at the BCR-ABL kinase—after initial cytoreduction of the elevated blast count—would be a reasonable initial approach to getting control of the disease. But without allogeneic transplant as consolidation, this would not offer me long-term control of the disease. So, more on transplant in a bit.

Standard induction for “fit” patients with AML has remained largely unchanged for several decades—until recently. The chemotherapy combination of 7+3 (7 days of cytosine arabinoside and 3 days of an anthracycline) has been the backbone of induction in the U.S. Consolidation chemotherapy with 3 to 4 subsequent cycles of high-dose cytosine arabinoside has been common practice. The development of a liposomal preparation containing a defined ratio of cytosine arabinoside and daunorubicin has shown increased efficacy in AML that arises from a pre-existing condition known as myelodysplastic syndrome (MDS). Also, drugs targeting specific mutations in genes such as FLT3, IDH1, or IDH2 are additional options that can now be used in combination with standard approaches—or alone in some instances. But perhaps most impressive has been the incorporation of the BCL2 inhibitor venetoclax in combination with azacytidine as a low-intensity regimen now approved by the U.S. Food and Drug Administration (FDA) for elderly and unfit patients with MDS.

Response rates appear to essentially equal those of the standard high-intensity induction regimens. Importantly, this regimen generally has lower toxicity and is most often administered outpatient. Recent data show that patients achieving a complete response with this regimen do as well with allogeneic transplant as folks who received more intensive induction. And for younger, fit patients, venetoclax added to more intensive regimens is producing results that are likely significant improvements over the old 7+3.

So, getting back to the first question I posed, am I looking at the possibility of a cure? Possibly. It is not likely, at my age, that cytogenetics would be favorable. Complex cytogenetics and AML arising from MDS are much more prevalent. Even if this were to be a Ph+ AML blast crisis, my long-term survival would depend on allogeneic transplant as consolidation post remission.

Am I a candidate at 70 for an allogeneic transplant? Possibly. Recent studies show that arbitrary age limits on allo transplantation have virtually disappeared across transplant centers. Less ablative regimens and improved supportive care around infection control and prevention of graft-versus-host disease have led to a situation where transplantation for older patients is a viable option with reasonable outcomes. Unfortunately, it is one that is currently afforded to only about 5% of older AML patients.

So, what would I want in this situation? I would, of course, look for any relevant clinical trials, as all advances in the field are dependent on trials of novel therapies. There are now several trials looking at the addition of novel targeted therapies as a component of induction therapy for patients with appropriate molecular features. But in the absence of an appropriate trial, for my intermediate- or poor-risk AML, I would likely opt for azacytidine and venetoclax as an induction/consolidation regimen, which would give me a better chance of escaping potentially debilitating complications of induction and consolidation that might put a definitive transplant out of reach. And, I would consider transplant as consolidation if in complete remission and still in good shape.

Some reality here: if these approaches didn’t get me to a complete remission, my discussions would center on defining the best way to preserve quality of life with family and friends during my remaining time.

Dr. Asch can be reached at adam-asch@ouhsc.edu.

Related links:

A message from Curious Dr. George:

The goal of Cancer Commons is to help patients identify and access their best possible treatments, one patient at a time, while moving the field forward. If you have advanced cancer, let our molecular oncology scientists provide personalized information about your options.

***

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

How Would an Expert Manage His Own Advanced Hodgkin Lymphoma: an Update

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

Kevin Knopf, MD Chief Medical Officer, Cadex Genomics; CMO and Division Chief of Hematology/Oncology, Highland Hospital (Oakland)

When facing a frightening new cancer diagnosis, some people ask their doctors, “What would you do if you were me?” Here, our Curious Dr. George asks oncologist Kevin Knopf, MD, how he would handle his own advanced Hodgkin lymphoma.

Curious Dr. George: Please consider this hypothetical scenario—as a very busy practicing clinical oncologist during the COVID-19 pandemic, you were not paying much attention to your own health when you noticed a weight loss of 7 pounds, a dry cough, and a little fever and sweating at night. Your home COVID-19 antigen tests were negative, but you found some enlarged cervical lymph nodes. Your physician confirmed the lymphadenopathy and palpated an enlarged spleen. Your hematocrit was 39, and WBC 16, 000. CT scan detected both para aortic and mediastinal lymphadenopathy. Biopsy of a cervical node was diagnosed as nodular sclerosing Hodgkin lymphoma. How do you proceed?

Kevin Knopf, MD, MPH: First, which chemotherapy regimen would I pick, given the treatment is with curative intent? Three standard choices: ABVD, BEACOPP, and A-AVD foster intense debate amongst oncologists. A recent Twitter poll by Dr. Aaron Goodman (of the University of California, San Diego) had opinions split evenly between ABVD, R-BEACOPP, and A-AVD. The choice has subtle nuances; my feeling is ABVD is probably fine for young patients where the risk of bleomycin pulmonary toxicity can be monitored closely; for myself I would choose A-AVD and monitor carefully for neuropathy.

Where to be treated? Where I work in the East Bay of California’s San Francisco Bay Area, there are centers for the University of California, San Francisco (UCSF) and Stanford, and Dr. Andreadis—a friend—is at UCSF, and Dr. Barbara Galligan, my former resident whom I helped mentor into oncology, are all excellent. By a thin margin I would choose Dr. Rajesh Behl in Berkeley in the East Bay. He is a smart oncologist and I appreciate his dry sense of humor. Getting in and out of his office for treatment would be easy, and it is with the infusion nurses that I would spend 98% of my time.

I am fortunate that my insurance would cover the complete cost and my choice of physicians, but this illustrates an area of oncology I spend a lot of time thinking and writing about: cost effectiveness and health equity. There are broad disparities in cancer care—within the U.S. and worldwide—such that many patients with a curable cancer, such as Hodgkin lymphoma, are not cured.

Pricing for cancer care seems arbitrary and is not really negotiated. In classic economics, one would pick amongst my 5 treatment options based on the lowest cost of care, as the outcome is almost certainly equal. There is a mind-blowing aspect of American health insurance: when Obamacare was enacted, insurance companies’ net profit was capped at 15% of the medical loss ratio, meaning the more my insurance company spends, the more money they can make. My premiums have jumped 15% in 2023. A-AVD is 108-fold more costly than ABVD. Cancer care in the U.S. is virtually “all accelerator, no brake,” and the cost of care keeps rising each year, and health disparities widen.

Sociologist C. Wright Mills quipped “the most important thing you can do in a capitalistic society is choose the right parents.” I chose wisely and ended up with “Cadillac” health insurance. All other industrialized nations have a single-party payer system to pay for care, while U.S. health care delivery is based on a for-profit model. American health care works out well in this particular hypothetical example, but overall, we rank about 50th in the world in terms of healthcare efficiency—tied with Bulgaria.

There are casualties. Half of the U.S. has medical debt, 50% of women with metastatic breast cancer are being pursued by debt collectors, and 42% of newly diagnosed cancer patients will go bankrupt within 3 years of diagnosis. We have a tragic and widening cancer disparity gap in the U.S. and a large care gap between first-world nations and low- and middle-income countries. In many parts of the world, a similar patient would not receive curative chemotherapy. People speak of the American health care “system,” but that would imply a formal interconnected design rather than our Byzantine way of paying for health care. So I might come out of this diagnosis just fine, and not bankrupt, but that was just a simple twist of fate.

Dr. Knopf can be reached at kevinbknopf@gmail.com.

***

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

SAGE Oncotest: Entering the Next Generation of Tailored Cancer Treatment

Chris Apfel, MD, PhD, MBA
Founder, CEO, and Board Chair of SageMedic Corp.

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

For people with recurring or drug-resistant cancer, choosing the best-possible next treatment can pose a huge challenge. Our Curious Dr. George asks Chris Apfel, MD, PhD, MBA—Founder, CEO, and Board Chair of SageMedic Corp.—about his company’s solution for many of these patients.

Curious Dr. George: Despite the application of curative surgical, radiation, and systemic treatments, cancer remains the “emperor of all maladies” with 600,000 Americans dying of cancer every year. With many more patients battling recurrent or resistant disease, even with adherence to standard guidelines from the National Comprehensive Cancer Network (NCCN), selecting the next line of treatment—whether cytotoxic chemotherapies, targeted therapies, or immunotherapies—often follows a trial-and-error approach.

To address this unresolved challenge, SageMedic, a CMS and CLIA-accredited and California-based company, has developed the SAGE Oncotest™. In comparison to next-generation genomic sequencing, which provides actionable insights for a smaller number of patients, SageMedic claims that its Oncotest, an extreme drug resistance (EDR) assay, provides actionable insights for virtually all patients within 10 days, thereby avoiding likely failures.

Dr. Apfel, I understand that this is only possible because you are using fresh, live cancer tissue, right? Can you share more about how your test works and how it can help cancer patients?

Chris Apfel, MD, PhD, MBA: The SAGE Oncotest is a next-generation EDR assay in which we use a patient’s biopsy specimen to create microtumors in the lab. We then expose the microtumors to a wide range of drug concentrations, and we measure which drugs at which concentrations are most effective at killing the cancer cells.

Within one day of receiving a fresh biopsy specimen, we can create hundreds of microtumors from virtually all types of solid cancer, including breast, ovarian, prostate, and pancreatic cancers, glioblastoma, and cancers of unknown origin. Thanks to our proprietary technology, these live microtumors maintain the patient’s tumor heterogeneity and microenvironment for virtually every biopsy specimen.

Furthermore, in contrast to organoid approaches that usually take weeks or months for organoid growth, if growth occurs at all, the SAGE Oncotest one-day aggregation step allows for next-day drug exposure, with actionable information generated for virtually every patient within just 7 to 10 days.

Our predicate EDR predicted tumor resistance with about 96% accuracy, and it is highly unlikely that a tumor will respond to a therapy that does not work ex vivo in supra-physiological concentrations, as in the context of the SAGE Oncotest. By ruling out ineffective therapies, we project this assay to identify the most promising drugs with about 80% accuracy, doubling a patient’s chance for a tumor response, significantly improving quality of life, and significantly extending the patient’s life expectancy.

SAGE can test the potential sensitivity and resistance of dozens of drugs and drug combinations, though our current technology does not test for anti-hormonal or immuno-oncology drugs.

If any readers are interested in having their patient’s tissue tested to help inform their treatment plan, we ask them to please let us know at least a week ahead of the patient’s biopsy or surgery, so that we can send the shipment kit to ensure overnight delivery of viable tissue. SAGE Oncotest requires fresh, live cancer tissue rather than formalin fixation, which kills cells within minutes and renders the sample unusable for our assay.

Physicians can choose from two Sage Oncotest panels. The standard panel (“Superior Care within Standard of Care”) tests the microtumors against NCCN guideline-recommended drugs and can inform, especially, initial treatment plans, e.g., stage II or III patients expected to require adjuvant therapy. The expanded panel (“Beyond Standard of Care”) includes the standard panel plus special requests, including targeted therapies and repurposed drugs, which becomes meaningful when standard treatment regimens fail. Of note, if a fresh biopsy is not feasible, we have also been able to retrieve sufficient cancer tissue from pleural effusions, ascites, and even bone metastases.

In summary, the SAGE Oncotest complements other precision oncology approaches by providing actionable information for virtually all patients within 7-10 days to avoid ineffective drugs and significantly increase the chance for best-possible outcomes.

Dr. Apfel can be reached at capfel@sagemedic.com.

***

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

Treating Pancreatic Cancer: Could Metabolism—Not Genomics—Be the Key?

Thomas Seyfried
Biology Department
science

Derek Lee, MS
PhD Candidate (Seyfried Lab), Department of Biology at Boston College

Tomás Duraj, MD, PhD
Postdoctoral Research Fellow, Department of Biology at Boston College

Curious Dr. George
Cancer Commons Editor in Chief George Lundberg, MD, is the face and curator of this invitation-only column

In 2018, we published a Curious Dr. George post featuring Thomas N. Seyfried, PhD, about cancer possibly being mostly a metabolic—rather than genetic—disease. Since then, the field of cancer genomics has expanded enormously, and we have published many posts in support of better understanding and treatment of many types of cancer based on the genomic approach. Here, Dr. Seyfried and two of his colleagues at Boston College—Derek Lee, MS, and Tomás Duraj, MD, PhD—provide an update on their perspective.

Curious Dr. George: Our recent Curious Dr. George post on pancreatic cancer stimulated you to push back on existing therapeutic strategies that have been failures for so long, and suggest an entirely different approach. Why do the current treatments of pancreatic cancer fail, and what new approach might be worth considering?

Thomas N. Seyfried, PhD; Derek Lee, MS; and Tomás Duraj, MD, PhD: Current therapeutic approaches for managing the most common form of pancreatic cancer (pancreatic ductal adenocarcinoma , or PDAC)—and many other cancers, for that matter—have failed to significantly reduce deaths in severe cases. The promise of effective precision medicine based on the somatic mutation theory of cancer has not been realized. This is evident from the latest statistics published by the American Cancer Society showing that, every day, almost 1,700 people die of cancer or cancer-treatment-related causes. While it is understandable and important to highlight recent advancements, the long-term (10-year) survival rate for PDAC is still less than 5%.

It is also important to be realistic about the side effects of many first-line treatments for PDAC. Many of the current “targeted” treatments can result in severe fatigue, gastrointestinal disturbances, liver failure, and myriad other complications. These adverse effects are unacceptable and contribute further to the physical and mental stress already associated with a cancer diagnosis. Treatment modalities based on an incorrect theory of cancer origin will not provide optimal management for most patients.

In contrast to the somatic mutation theory, the mitochondrial metabolic theory can better explain the origin of cancer and provide more rational therapeutic strategies for management. For example, KRAS mutations negatively impact oxidative phosphorylation (OxPhos), thus forcing tumors to be more dependent on aerobic fermentation than on OxPhos for driving dysregulated growth. OxPhos inefficiency coupled to energy synthesis through aerobic glucose and glutamine-dependent fermentation is now recognized in nearly all cancers. Unfortunately, the metabolic dependencies of cancer cells are just an afterthought, if considered at all.

We have yet to find a report showing any cancer cells that can grow in the absence of glucose and glutamine. Ketone bodies and fatty acids are non-fermentable fuels and cannot therefore replace either glucose or glutamine for sufficient energy production in cancer cells with OxPhos insufficiency. An effective non-toxic therapeutic strategy for managing cancer would therefore involve the simultaneous down-regulation of the glucose-driven glycolysis and the glutamine-driven glutaminolysis pathways using diet-drug combinations, while transitioning the body to non-fermentable fuels, e.g., fatty acids and ketone bodies.

A transition of the body from glucose to nutritional ketosis—glucose/ketone index (GKI) values of 2.0 or below—will induce significant physiological stress on the already metabolically inflexible cancer cells while, at the same time, providing an efficient energy source for healthy cells that evolved to transition from glucose to ketone bodies under dietary energy reduction. A combination of both carbohydrate restriction and high-intensity exercise will stimulate glucose and glutamine clearance from circulation.

Adherence to this approach can be challenging for some, but shifting the role of the patient from passive observer to active participant in their non-toxic metabolic treatment strategy will help enable longer-term management of their cancer.

Dr. Seyfried can be reached at thomas.seyfried@bc.edu, Dr. Lee at leebig@bc.edu, and Dr. Duraj at tomas.duraj@bc.edu.

Additional references for successful application of ketogenic metabolic therapy in animals and humans:

İyikesici MS, Slocum AK, Slocum A, Berkarda FB, Kalamian M, Seyfried TN.Efficacy of Metabolically Supported Chemotherapy Combined with Ketogenic Diet, Hyperthermia, and Hyperbaric Oxygen Therapy for Stage IV Triple-Negative Breast Cancer. Cureus. 2017 Jul 7;9(7):e1445. doi: 10.7759/cureus.1445.

Elsakka AMA, Bary MA, Abdelzaher E, Elnaggar M, Kalamian M, Mukherjee P, Seyfried TN. Management of Glioblastoma Multiforme in a Patient Treated With Ketogenic Metabolic Therapy and Modified Standard of Care: A 24-Month Follow-Up. Front Nutr. 2018 Mar 29;5:20. doi: 10.3389/fnut.2018.00020.

Augur ZM, Doyle CM, Li M, Mukherjee P, Seyfried TN. Nontoxic Targeting of Energy Metabolism in Preclinical VM-M3 Experimental Glioblastoma. Front Nutr. 2018 Oct 5;5:91. doi: 10.3389/fnut.2018.00091. eCollection 2018.

Mukherjee P, Augur ZM, Li M, Hill C, Greenwood B, Domin MA, Kondakci G, Narain NR, Kiebish MA, Bronson RT, Arismendi-Morillo G, Chinopoulos C, Seyfried TN. Therapeutic benefit of combining calorie-restricted ketogenic diet and glutamine targeting in late-stage experimental glioblastoma. Communications Biol. 2019 May 29;2:200. doi: 10.1038/s42003-019-0455-x.

Seyfried TN, Shelton L, Arismendi-Morillo G, Kalamian M, Elsakka A, Maroon J, Mukherjee P. Provocative Question: Should Ketogenic Metabolic Therapy Become the Standard of Care for Glioblastoma? Neurochem. Res. 2019 Apr 25. doi: 10.1007/s11064-019-02795-4.

Seyfried TN, Mukherjee P, Iyikesici MS, Slocum A, Kalamian M, Spinosa JP, Chinopoulos C. Consideration of Ketogenic Metabolic Therapy as a Complementary or Alternative Approach for Managing Breast Cancer. Front Nutr. 2020 Mar 11;7:21. doi: 10.3389/fnut.2020.00021.

İyikesici MS, Slocum AK, Winters N, Kalamian M, Seyfried TN. Metabolically Supported Chemotherapy for Managing End-Stage Breast Cancer: A Complete and Durable Response. Cureus. 2021 Apr 26;13(4):e14686. doi: 10.7759/cureus.14686.PMID: 33927959.

Seyfried TN, Shivane AG, Kalamian M, Maroon JC, Mukherjee P, Zuccoli G. Ketogenic Metabolic Therapy, Without Chemo or Radiation, for the Long-Term Management of IDH1-Mutant Glioblastoma: An 80-Month Follow-Up Case Report. Front Nutr. 2021 May 31;8:682243. doi:10.3389/fnut.2021.682243. eCollection 2021.PMID: 34136522.

Seyfried TN. The effects of diet on prostate cancer outcomes. Nat Rev Urol. 2022 Jul;19(7):389-390. doi: 10.1038/s41585-022-00612-2.PMID: 35676538.

 

***

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.