Tag Archive for: clinical trials

Peer Insights: Fostering Clinic-Wide Engagement for Myeloma Clinical Trials

Peer Insights: Fostering Clinic-Wide Engagement for Myeloma Clinical Trials from Patient Empowerment Network on Vimeo.

Dr. Craig Cole from Karmanos Cancer Institute speaks to the success of clinical trials relying on not only provider endorsement, but also on the collective enthusiasm and involvement of all healthcare staff, which cultivates a patient-centric culture promoting myeloma trial participation and engagement.

See More from EPEP Myeloma

Related Resources:

Peer Insights: Maximizing Myeloma Patient Care

Peer Insights: Maximizing Myeloma Patient Care 

HCP Strategies for Navigating the Pre-trial Eligibility and Informed Consent Process

HCP Strategies for Navigating the Pre-trial Eligibility and Informed Consent Process 

How Can Myeloma HCPs Overcome Unforeseen Practice Related Barriers?

How Can Myeloma HCPs Overcome Unforeseen Practice Related Barriers? 

Transcript:

Dr. Craig Cole:

Some of the other barriers to clinical trials, the nurses and the other staff in the cancer center aren’t aware of the clinical trials, that when a patient goes through the clinic, they talk to more than just the provider. They talk to the treatment nurses, they talk to the intake people, they talk to the MAs, they talk to the scheduling people. And there was a study that was done a few years ago in looking at patients who were given consent forms and declined clinical trials. And they found that a lot of patients declined clinical trials, were because they said that, well, their doctor didn’t want them on the trial.

And when they looked further into that, they saw that, well, the doctor offered them a clinical trial, but when they discussed the clinical trial with a nurse practitioner, when they discussed that trial with a treatment nurse or the MA or any of the other staff, when they didn’t know about the clinical trial, that was considered well, if you don’t know about the clinical trial, it must not be good for me. And then they withdrew from the trial.

It really shouldn’t be left in the provider compartment. That excitement should be clinic-wide. And when you have that all-in approach where everybody’s involved, everyone’s excited about clinical trials, it produces a culture of clinical trials that everybody wants to be part of, and the patients then can jump on that bus and feel comfortable participating in the trial. 

Advanced Prostate Cancer Treatments on the Horizon

Advanced Prostate Cancer Treatments on the Horizon from Patient Empowerment Network on Vimeo.

 What advanced prostate cancer treatments are on the horizon? Expert Dr. Ronald Chen discusses emerging treatments that are under study, the advantages that the treatments might offer, and how patients can potentially gain access to the therapies.

Download Resource Guide

See More from START HERE Prostate Cancer

Related Resources:

How Is Advanced Prostate Cancer Explained to Newly Diagnosed Patients?

How Is Advanced Prostate Cancer Explained to Newly Diagnosed Patients?

How Can Prostate Cancer Collaborative Care Be Coordinated?

How Can Prostate Cancer Collaborative Care Be Coordinated?

START HERE | Collaborative Prostate Cancer Care Resource Guide 

Transcript:

Lisa Hatfield:

Are there any promising treatments on the horizon that you are particularly excited about for advanced prostate cancer? And what types of questions do you recommend patients and their care partners ask of their providers?

Dr. Ronald Chen:

I want to answer this question from the perspective of a…as a radiation oncologist. I am often involved in a lot of research and clinical trials related to how we can potentially better use the tool of radiation for patients with prostate cancer and advanced prostate cancer. And there’s actually quite a bit of exciting development in radiation that I think I’m really looking forward to seeing the results of those types of research. One area of, I think, promising treatment is something called radiopharmaceutical therapy.

So oftentimes, we think of radiation as a patient…there’s a machine that delivers a targeted beam of radiation to a particular tumor. And that’s what we usually think about. But actually, there’s a new wave of radiation and how we deliver it called radiopharmaceuticals. And radiopharmaceuticals is almost like getting chemotherapy. It’s something that’s infused into the bloodstream, almost like chemotherapy, but the way this treatment work called radiopharmaceuticals is that you’re actually infusing molecules that will actually tag on to tumor cells in the body, and then as it tags onto tumor cells in the body, deliver radiation to that tumor cell.

And they’re actually…so actually, it’s kind of like chemotherapy delivering actually radiation instead of a drug. And there are actually already are several of these radiopharmaceutical agents that have been proven to be effective. And two of these have already been FDA approved that are now in use for prostate cancer. And we know they work and they extend survival. And so, I think that’s very exciting.

And one of them is called lutetium Lu 177 vipivotide tetraxetan (Pluvicto). You’re infusing a molecule that specifically tags on to prostate cancer cells in the body and delivers a little bit of radiation to that cell. And that extends survival for patients. It’s actually a very well-tolerated treatment as well as actually now commonly in use FDA-approved. And I think that now that we have a couple of these FDA-approved, they were proven to work, I really do think that this field of radiopharmaceutical treatment will continue to expand. There’ll be more and more of these in the future. There’s more in clinical trials, and I think there’ll be more and more available options in the future. And I think it’s really going to be a great way to potentially use radiation to help patients with advanced prostate cancer and other cancers in the future.

Another thing that I’m really excited about is, again, I think the balance between extending a patient’s life expectancy and balancing the quality of life impact. We know that for patients with advanced prostate cancer, a common way to treat this disease is with hormone therapy. And oftentimes, people are on hormone therapy for years and years and maybe lifelong. And we also know that hormone therapy in patients with prostate cancer can also have really quite a bit of side effects. It can make people fatigued, weight gain, it may have cardiovascular disease impact. And so hormone therapy, even though it’s effective for prostate cancer, really has a big quality of life impact on patients.

So one of the current years of research and in clinical trials is, can we, instead of having patients with metastatic prostate cancer, instead of having them on hormone therapy for life, could we potentially use radiation selectively in the spots of metastasis? And if we’re able to use radiation to treat particular spots of metastasis and that’s controlled, can we let the patient then have a break from hormone therapy, which I think would really have an important quality of life improvement.

And so selectively using radiation to treat a few spots, allowing patients a break from hormone therapy is another area of research. There’s actually a couple of clinical trials already done on this. And what it’s shown so far is that if patients have one to three to five, a few spots of metastasis, using radiation can actually give patients a break from hormone therapy for two, three or four years for many patients. And that can really have a major impact on improving quality of life without compromising their survival outcomes. So I think I’m really excited about those kinds of areas of research.

One more treatment option like radiopharmaceuticals, two treatments that not compromise the patient’s survival, but improve quality of life. I’m excited about those directions. Oftentimes, the new developments and new treatments, the promising treatments are first available through clinical trials, and only through participating in clinical trials do you have access to that before it becomes FDA-approved. And oftentimes, that could be a really good option to consider. So that’s why I think it’s really important to ask that for every patient.

Advancements in Endometrial Cancer Trials | Insights and Opportunities

Advancements in Endometrial Cancer Trials: Insights and Opportunities from Patient Empowerment Network on Vimeo.

What’s important for endometrial cancer patients to know about clinical trials? Expert Dr. Charlotte Gamble from MedStar Health discusses novel therapies under study, treatments that are showing promising results, and patient advice on clinical trials.

[ACT]IVATION TIP

“…be able to ask your doctor about if you’re eligible for clinical trials, what your cancer mutational or genetic code is that might make you eligible for certain clinical trials, and where those trials are offered, if it’s at the health system that you are seeking care, or if it’s at a nearby health system, if you’re able and willing to travel.”

Download Guide | Descargar Guía en Español

See More from [ACT]IVATED Endometrial Cancer

Related Resources:

What Are Barriers to Endometrial Cancer Care Access?

What Are Barriers to Endometrial Cancer Care Access?

Should Some Gynecologic Cancer Patients Seek a Specialist?

Should Some Gynecologic Cancer Patients Seek a Specialist?

Are Beauty Products a Risk Factor for Endometrial Cancer?

Are Beauty Products a Risk Factor for Endometrial Cancer?

Transcript:

Lisa Hatfield:

So, Dr. Gamble, this is kind of a three-part question about clinical trials. Can you talk a little bit about ongoing clinical trials that are investigating novel therapies for advanced endometrial cancer? And then maybe talk a little bit about some promising or encouraging results that you’re seeing with trials? And last part of the question is, what do you want people living with endometrial cancer to know about clinical trials?  

Dr. Charlotte Gamble

I’m so glad you asked this question. This is such a valuable question and an area of a lot of interest that has improved over the past several years, not only about clinical trials and the real need to make sure that patients are aware of them and to ensure that these clinical trials represent the populations that we intend to serve as healthcare providers in the United States, but also specifically in the world of endometrial cancer. Really exciting, promising results that we’ve had over the past year, year-and-a-half specifically that address the very desperate need for novel therapeutics to treat patients who have endometrial cancer.

So, for example, two major trials were published last year, presented at international meetings, looking at the real improvement in overall survival, really increasing the length of time patients can live with endometrial cancer that leverage the use of drugs called immunotherapy. So things like dostarlimab-gxly (Jemperli) or pembrolizumab (Keytruda), these are generic names for immunotherapy drugs that work very well in some subsets of patients with endometrial cancer.

This is something, some survival benefits that we have never seen before in the endometrial cancer space and rarely seen in the gynecologic oncology space and is a definite marker of huge success in terms of extending the lifespan of patients who suffer from this challenging to treat understudied, underfunded disease. Endometrial cancer is actually one of the lowest funded studies in the National Cancer Institute at NIH.

And so having major trials come out over the past couple of years that really look at survival opportunities with the leveraged use of immunotherapies is something that is both exciting and invigorating to the field and hopefully can potentiate further funding from the NCI to be able to study this disease type. In terms of your question for what patients should know about, about ongoing trials, I think this dovetails into several of the points that we’ll discuss during this interview of making sure that patients are their own advocate and having an advocate nearby and with them at all of their appointments.

 So it’s really important to ask their subspecialists, their oncologists or their gynecologic oncologists about if there are any clinical trials that the patients are eligible for. A lot of this comes down to, has the patient undergone genetic testing or molecular sequencing that looks at the specific mutations in the cancer tissue that sometimes will make patients eligible for certain clinical trials or others? And other times it’s just understanding that what opportunities are available within the health system and outside the healthcare system in which the patient is seeking care.

A lot of times when we see that these trials that are published might not represent a racially diverse group of patients. Oftentimes it’s because of two reasons. One, patients aren’t even offered clinical trials, even if they are eligible. Or two, patients might be getting care at a health facility that doesn’t have access or the infrastructure to enroll them on these clinical trials that could be available, perhaps at a regionally nearby cancer center.

I oftentimes suggest to patients, please ask me questions about your molecular subtyping. Ask me questions about what clinical trials you might be available for. There is a significant amount of trust that the health system needs to earn back from patients to allow them the headspace to trust the health system again, given historical, massive, ethical issues and trials in the past and patients and their loved ones feeling that clinical trials just means a big experiment and they don’t want to be experimented on. And what I often say to that is really, you have to understand the details of the trial and the science going into it and make sure that your doctor has your best interests at heart. But oftentimes these trials hold significant promise.

And the reason that you might be eligible for them is that the trial drugs might work better than standard of care, certainly for endometrial cancer we’ve seen that in the two major trials that came out this year. So I think my activation tip for this question is really to be able to ask your doctor about if you’re eligible for clinical trials, what your cancer mutational or genetic code is that might make you eligible for certain clinical trials, and where those trials are offered, if it’s at the health system that you are seeking care, or if it’s at a nearby health system, if you’re able and willing to travel.


Share Your Feedback:

Create your own user feedback survey

How Can Myeloma Patients Access CAR T-Cell Therapy Clinical Trials?

How Can Myeloma Patients Access CAR T-Cell Therapy Clinical Trials? from Patient Empowerment Network on Vimeo.

How can patients learn more about joining myeloma CAR T-cell therapy clinical trials? Dr. Krina Patel shares advice for identify and accessing these trials, noting that seeking care with a myeloma specialist can be most helpful. 

Dr. Krina Patel is an Associate Professor in the Department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center in Houston, Texas. Dr. Patel is involved in research and cares for patients with multiple myeloma.

Related Resources:

Will CAR T-Cell Therapy Be Approved for Earlier Lines of Myeloma Treatment

Will CAR T-Cell Therapy Be Approved for Earlier Lines of Myeloma Treatment?

How Is CAR T-Cell Therapy Research Advancing Myeloma Care?

How Is CAR T-Cell Therapy Research Advancing Myeloma Care?

Understanding Myeloma Therapy Targets BCMA and GPRC5D

Understanding Myeloma Therapy Targets BCMA and GPRC5D

Transcript:

Katherine:

How can patients find and access clinical trials that are looking at CAR T-cell as an earlier line of therapy?  

Dr. Krina Patel:

That’s a great question. So, I think going to any place that is myeloma specific. So, basically a big center that has doctors that are doing myeloma research, they will be able to definitely get you into places that have some of these trials. But clinicaltrials.gov is one other place. It’s really hard. I will tell you that, if I wasn’t a physician or in medicine, I don’t think I would learn, I would be able to navigate it very well.  

And so, really either through your doctor and having them look this up for you, or going to patient groups. So, again, a lot of my patients are part of different patient groups where people will say, “Well, this is a trial that I was” or “This is a trial that my doctor told me about.” And then, asking. So, that’s the other big thing is constantly asking your doctor “What are my other options?” getting second opinions from myeloma experts, and then just paying attention to some of these resources that you have available. Right now, there are gonna be more clinical trials for earlier-line therapies and first-line with both cilta-cel and ide-cel 

There’s going to be clinical trials with new products: ddBCMA CAR T, that is likely gonna come out soon for earlier-line therapies. And so, there’s a lot happening, and so there might be different clinical trials in different places. But I think the fact that all this is going on at once is really important for our patients to know about. 

Available Myeloma Treatment Options for Patients | An Overview

Available Myeloma Treatment Options for Patients | An Overview from Patient Empowerment Network on Vimeo.

What are the current myeloma treatment approaches? Myeloma expert Dr. Sikander Ailawadhi shares an overview of treatment options, the necessity of combination therapy in myeloma, and the role of clinical trials in patient care. 

Dr. Sikander Ailawadhi is a hematologist and oncologist specializing in myeloma at Mayo Clinic in Jacksonville, Florida. Learn more about Dr. Ailawadhi.

Related Resources:

Key Advice for Myeloma Patients | Questions to Ask About a Care Plan

The Benefits of Shared Decision-Making for Myeloma Care

The Benefits of Shared Decision-Making for Myeloma Care

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care?

What Is the Role of Bispecific Antibody Therapies in Future Myeloma Care? 

Transcript:

Katherine:

What are the types of treatments available for people with myeloma?   

Dr. Ailawadhi:

So, myeloma has a lot of treatments available.  We can classify these treatments into different classes of drugs, or we can classify the treatment as early lines or late lines of therapy. Or we can classify these treatments into cellular therapy or targeted therapy or chemotherapy. There are ways of classifying it.  What I would suggest is we should think about classes of drugs.  We have something called proteasome inhibitors. That class has three drugs FDA-approved. We have something called immunomodulatory drugs. That class has three drugs also approved, but generally, we use two.   

Then, there are something called monoclonal antibodies. There are three drugs approved there as well.    

There are cellular therapies or CAR T-cell therapy. There are two of them approved.  There is also a stem cell transplant that is used as a part of treatment sometimes but is different from CAR T. Then, there are other immunotherapy, something called T-cell engagers, in which also there are three drugs approved. In fact, as I’m saying to you, I’m trying to think…yeah, wow. Every class has three drugs. That’s so weird. And then, there are some other classes of drugs. There is something called exporting inhibitors. There is a drug there. All said and done, there are these different classes of drugs.  

There are some guiding principles for myeloma treatment. Generally, three to four drug combinations or regimens are better than two drugs. So, a patient should be in the initial therapy or later lines. Also, preferably be getting a three-drug combo. And I forgot to mention steroids, which are an important part of every regimen in myeloma, almost every regimen. So, three drugs or four drugs are better than two. That’s important to keep in mind. Longer durations of treatment are generally considered better.  We should not tinker with the regimen’s recipe too soon. As long as the patient is tolerating for a longer duration before making any major changes like maintenance.  

Generally, maintenance in myeloma is not a response-assessed thing like, “Oh, you’ve responded in two months. We should go to maintenance.” Generally, in myeloma, maintenance transition is a time-dependent thing. Okay, you’ve had six or nine months or 12 months. We can go to maintenance, sort of a thing. So, even if somebody has responded, they may need the same treatment for a longer period of time to keep the disease quiet.  

And so, I think these are the different categories of drugs. We pick and choose from different categories to combine and make a regimen. The CAR T-cell therapy, the two CAR Ts that are approved, or the three T-cell engagers that are approved, they are all currently used as single agents. They are not combined with anything, not even with steroids. 

Katherine:

How do clinical trials fit into a treatment plan?    

Dr. Ailawadhi:

Okay, that’s an extremely important question, and you’re asking it from a person in my clinic about two-thirds of the patients who are on treatment at any given time are on clinical trials. So, I am very heavily, I shouldn’t say biased, but a proponent of clinical trials. In my opinion, clinical trials are a part and parcel of treatment for every single patient. In fact, when you look at the NCCN guidelines, which are National Comprehensive Cancer Network, which is large institutions across the country, and they make guidelines for all cancers, it is mentioned in every single setting that clinical trials should always be considered.  

So, I personally feel that whenever the patient is coming up with a treatment decision, we talked about shared decision-making in the beginning, it’s important for them to ask at every single juncture, “Do you have any clinical trials available for me? 

And if you don’t have any clinical trials available, are there any clinical trials that I should consider, even if it means going to a different place and getting an opinion?” I know logistically it’s challenging, but we should at least know our options. So, in my opinion, clinical trials should be considered at every single juncture, because that is how patients get access to either a new drug, a new treatment, or a different way of using the current drugs, which might actually improve upon their current state. So, everybody all the time should consider clinical trials.    

Advances in Managing CAR T-Cell Therapy Side Effects

Advances in Managing CAR T-Cell Therapy Side Effects from Patient Empowerment Network on Vimeo.

What progress is being made in treating the side effects of CAR T-cell therapy? Myeloma expert Dr. Krina Patel discusses the research and advances being made in understanding and managing the common issues associated with this treatment.

Dr. Krina Patel is an Associate Professor in the Department of Lymphoma/Myeloma at The University of Texas MD Anderson Cancer Center in Houston, Texas. Dr. Patel is involved in research and cares for patients with multiple myeloma. 

See More From Thrive CAR T-Cell Therapy

Related Resources:

CAR T-Cell Therapy for Myeloma | What Are the Advantages

CAR T-Cell Therapy for Myeloma | What Are the Advantages?

Recovering From CAR T-Cell Therapy | What Can Myeloma Patients Expect

Recovering From CAR T-Cell Therapy | What Can Myeloma Patients Expect?

What Side Effects Are Possible Following CAR T-Cell Therapy?

What Side Effects Are Possible Following CAR T-Cell Therapy?

Transcript:

Katherine Banwell:

Dr. Patel, what about managing the side effects of CAR T? Is that improving?  

Dr. Krina Patel:

It is. I think just the fact that we are now seeing less of the neurotoxicity that we were seeing in the initial patients that were getting CAR T, and the serious neurotoxicity like the Parkinsonianism or patients couldn’t walk or hold things because of the tremors they were having that were so significant.  

And then something kinda like Guillain-Barré where people are getting ascending paralysis. And so, again, those are very serious things that we don’t want any of our patients to get. And again, most of it was with cilta-cel and their CARTITUDE-1 study. There is a black-box warning with ide-cel too that that can happen.  

We just don’t see it at the same rate. And again, when patients got better bridging going into CAR T, that rate dropped from 6% to 0.5%. So, it really is a huge difference by just giving better bridging and having myeloma controlled better going in.  

We are seeing some other side effects that we didn’t necessarily see on the original clinical trials, like facial palsies. These are things where patients can talk very well anymore or their side of their face is drooping. And those are reversible thankfully, for the most part with steroids, things like that. So, we watch for that. There’s some other side effects.

Again, these are immune cells going into your body, so anything can happen. So, we definitely keep a close eye on all of our patients. But for the most part, especially things like infections and this neurotoxicity, we’ve learned that again the less myeloma going in, the better patients do and the faster they recover.  

So, for the most part, patients now are doing much, much better than when patients were originally on those trials where we didn’t know what was gonna happen. And I think the infection piece is such a big deal; that our patients definitely need supportive care.  

So, even though this is a one-and-done where you get your chemo, you get your CAR T’s and then you’re not on any myeloma therapy, you are on supportive therapies; so, things like IVIG for your immune system, things to protect you against something called PJP pneumonia for at least six months if not longer.  

You’re still on anti-shingles medication. So, it’s not that you stop all medicines, but it’s you stop the myeloma medicines. And then, at least for the first six months, maybe even up to a year we have to make sure you don’t get any major infections.  

PODCAST: Kidney Cancer Patient Expert Q&A: Dr. Moshe Ornstein


START HERE bridges the expert and patient voice, enabling patients facing a kidney cancer diagnosis to feel comfortable asking precise questions of their healthcare team.

In this program, Dr. Moshe Ornstein offers invaluable insights into renal cell carcinoma (RCC), equipping newly diagnosed patients with essential information on treatment priorities, as well as strategies for managing progression and recurrence.

Download Resource Guide

See More from START HERE Renal Cell Carcinoma (RCC)

Transcript:

Lisa Hatfield:

Hello and welcome. My name is Lisa Hatfield, your host for this Patient Empowerment Network START HERE program, where we bridge the expert and patient voice to enable you and me to feel comfortable asking questions for our healthcare team. The world is complicated, but understanding your kidney cancer doesn’t have to be. The goal of this program is to create actionable pathways for getting the most out of treatment and survivorship. 

Joining me today is Dr. Moshe Ornstein. Dr. Ornstein is a respected kidney cancer physician and researcher from Cleveland Clinic. Dr. Ornstein’s clinical practice and research are focused on cancers of the genitourinary system, specifically kidney, bladder, and prostate cancer. He has published multiple research articles and presented his research at a variety of national medical meetings. He’s actively involved in multiple clinical trials. Dr. Ornstein, it’s such a pleasure having you today, and thank you for joining us. 

Dr. Moshe Ornstein:

Thanks for having me, Lisa. Pleasure to be here.

Lisa Hatfield:

So before we dive into today’s discussion, please take a moment to download the program resource guide using the QR code. This guide contains pertinent information to guide you both before and after the program. In this program, we’ll provide you with a comprehensive update on the latest kidney cancer news, and its implications for you and your family. Following that, we’ll launch into questions we have received from you.

So let’s start here. Dr. Ornstein, the treatment landscape of renal cell carcinoma, also known as kidney cancer, has evolved significantly over the past three decades, leading to improved therapeutic options and prolonged survival. That said, we also recognize that there are unmet needs to improve outcomes in kidney cancer. Today, we are privileged to have your expertise to help us decipher these developments and shed light on the advancements shaping the landscape of kidney cancer care. Dr. Ornstein, can you speak to the latest news in renal cell carcinoma?

Dr. Moshe Ornstein:

Sure, Lisa. It’s a great question. But before I start with the most recent updates and the latest news, I just want to clarify something. I think for you, Lisa, as you say the words kidney cancer and renal cell carcinoma, it’s indeed a mouthful. And for our patients, it can be very confusing as they sometimes hear kidney cancer, they sometimes hear renal cell carcinoma, they sometimes hear RCC, and it can be really hard to digest and understand what their diagnosis is, or if they see a presentation or a talk on kidney cancer, what that means.

So kidney cancer just means any cancer that arises in the kidney. There’s lots of different types of kidney cancer. For the purposes of today’s discussion, we’re really talking about renal cell carcinoma or RCC, which is the most common type of kidney cancer. And within RCC, the most common subtype is clear cell renal cell carcinoma.

So today, when you hear us talk about kidney cancer, RCC, renal cell carcinoma, clear cell RCC, just know that we’re referring to the most common type of kidney cancer. But it is a really important thing to talk to your doctors about, and for patients to talk, and their families to talk to physicians about to understand the subtype. We’ll use colloquially kidney cancer and RCC for today’s discussion. So just by way of a 30,000 foot view, when it comes to renal cell carcinoma, approximately two-thirds of patients are diagnosed when the cancer is in a localized stage, where the cancer is treated with curative intent, generally with surgery.

For patients who present with metastatic kidney cancer,  in other words, kidney cancer that has spread beyond the kidney, or for patients who had their kidney removed and then developed a recurrence or the cancer had come back to the lungs or the bones or anywhere beyond the kidney, those patients are treated with what’s called systemic therapy. Those are medicines that really cover head to toe, not a specific area, but head to toe.

And when we think about treatment options in kidney cancer, there are two main treatment options. One is immunotherapy. Immunotherapy is generally what’s called checkpoint inhibitors. And these are therapies that “release the brakes” on the immune system, and allow the body’s immune system to be activated to target the cancer. And the other type of medicine is called targeted therapies. And these for the most part, are vascular targeted therapies, and the way I describe it is they shrink the blood supply to the tumors. So again, you have those tumors that are diagnosed at a local stage. You have those tumors that are metastatic or advanced beyond the kidney. And the main treatment paradigms have to do with immunotherapy and targeted therapy.

So we just had the ASCO GU meeting, and I just want to describe the updates and how they fit into sort of the overall treatment paradigms across the different treatment sections, in other words, localized and metastatic. So for a patient who presents and comes in with just a kidney mass, that’s a kidney cancer, generally that patient’s going to be treated with surgery.

In general, there’s no rule for therapy before surgery. For many years, for that patient who had their kidney cancer removed from the kidney, either part of the kidney or the whole kidney removed, we really didn’t know what to do with those patients, and the standard of care was just to watch those patients. And we’ll get into a discussion about what watching the patient means. But one of the updates from the recent meetings has been that for patients who have their kidney removed because of kidney cancer, there is now a rule in some patients, this has to be a discussion with the doctor, to use immunotherapy to help prevent or delay the cancer from coming back. It’s a personal discussion.

We have a lot of data to support the use of a medicine called pembrolizumab (Keytruda), which is an immunotherapy that patients would get for a year after their kidney surgery. So that’s really the big recent update in the localized kidney cancer world, where the kidney cancer has been removed by surgery, and there’s now a treatment option, a year of immunotherapy after surgery for the right patient. So now, we move to the metastatic patient.

So again, the patient who has metastatic disease, either comes in with metastatic disease upfront, meaning the kidney’s there, the tumor’s in the kidney, and there’s advanced disease. And the other type is the patient who had their kidney removed a year ago, two years ago, sometimes five years ago, and now shows up with new spots in the lungs or the bones or elsewhere in the body. And that is metastatic or advanced kidney cancer.

So by in large, the overwhelming majority, and in my clinical practice, 95 percent of these patients are going to get an immunotherapy-based regimen as the first treatment for advanced kidney cancer. And there are different types of immunotherapy-based regimens. There’s an immunotherapy in combination with immunotherapy, and that’s called ipilimumab (Yervoy) and nivolumab (Opdivo), so double immunotherapy, or an immunotherapy plus a targeted therapy.

Lisa, we spoke about the targeted therapy cutting the blood supply. So in addition to getting two immunotherapies, some patients won’t get two immunotherapies, they’ll get one immunotherapy in combination with a targeted therapy. And those combinations include axitinib (Inlyta) and pembrolizumab, lenvatinib (Lenvima) and pembrolizumab and cabozantinib (Cabometyx) and nivolumab as the primary combination treatments for the first line of therapy for metastatic kidney cancer.

And the real updates from the recent meetings in this setting is just that with additional follow-up, in other words, we’ve seen follow-up at two years after the trial started, three years, four years, now five years, we’re seeing that there’s a subset of patients that continue to benefit with this combination years down the road. So, encouraging for patients. Again, it’s not every patient, different patients need different things, but just the knowledge that we have long-term follow-up data for patients who have gotten an immunotherapy-based combination for the front-line treatment for their advanced kidney cancer.

And the last update I want to touch on is once we move beyond that first line of immunotherapy-based combinations, we really don’t know exactly what to do beyond that. Meaning, if somebody got an immunotherapy-based combination, and then the kidney cancer got worse, what do we give next? And generally, we’re giving more of these vascular inhibitors, these targeted therapies. And the latest advancement in this area, in the refractory setting, in other words post immunotherapy-based combination is the introduction of a new medicine called belzutifan (Welireg), which is not a classic vascular inhibitor, but is something called the HIF-2α inhibitor. It’s a very well-tolerated therapy in many of the patients. And it does have activity in the right patient. And it’s now FDA-approved relatively recently for patients who have already had an immunotherapy-based combination. So that’s kind of the major update.

The post-surgery treatment with immunotherapy, long-term data for immunotherapy-based combinations in the metastatic setting, and a novel therapy, a new mechanism of action with a pill with a therapy called belzutifan for patients whose kidney cancer is getting worse despite standard treatments upfront.

Lisa Hatfield:

Okay. Thank you so much for that outstanding overview. I had one follow-up question to that overview regarding clinical trials. So can you talk about any clinical trials you are excited about, both in the newly diagnosed setting or in the…I can’t remember what you called it, but the newly diagnosed setting and then the metastatic or recurring setting for kidney cancer, specific clinical trials, and then some of the medications that you had mentioned, are those FDA-approved right now or are those also in clinical…are most of those in clinical trials at the moment?

Dr. Moshe Ornstein:

A great question. I think what this goes to show is that, here I am talking to you, and sometimes some of the words are hard to understand. You can imagine a patient with a newly diagnosed kidney cancer, how confusing a lot of this can be. So I’m really happy we’re having this discussion. Everything I had mentioned up until this point is FDA-approved. And if I am to mention something that is not FDA-approved, I’ll make that caveat. And while we’re talking about non-FDA-approved therapies, let’s talk about some of those new and exciting clinical trials. The way I look at clinical trials, whether it’s in the treatment naive, so a patient who has a newly diagnosed cancer, or in a patient with refractory cancer, meaning cancer that has gotten worse despite some treatment already.

So I look at clinical trials, and I tend to divide them into two different main categories. And I think for patients, this sort of helps categorize them in a neat fashion. One, is looking at those trials that are investigating novel therapies. So we spoke about those immunotherapy checkpoint inhibitors, we spoke about those vascular endothelial growth factor inhibitors, in other words, targeted therapy. We spoke about this HIF-2α, those are all therapies that we understand the mechanism of action, we understand how they work.

So one kind of clinical trial is saying, what’s next? What’s down the road? What’s not an irregular immunotherapy? What’s not a regular targeted therapy? What’s not another HIF-2α drug? What are the novel therapies being investigated? So some of those trials that I’m interested in are trials that are looking at something called bispecifics. So these are singular drugs that sort of have two different targets. We’re looking at cellular therapies. We know these things called CAR-T cells work well in some of other cancers like lymphomas, but is there a rule for using this type of novel mechanism in kidney cancer?

Drugs looking at things called antibody drug conjugates, which again, these types of therapies are available in breast cancer, in bladder cancer, in other types of cancer, but not yet in kidney cancer. And that’s kind of the one category of novel mechanisms, novel agents. That’s one class of clinical trials. And the other class of clinical trials is really sort of optimizing the drugs we already have. So we know that as a general rule, giving immunotherapy plus targeted therapy is better than giving immunotherapy alone. But what about trials looking at giving two immunotherapies plus a targeted therapy?

We know that patients either get immunotherapy and immunotherapy, or an immunotherapy and a targeted therapy. What about if we gave two immunotherapies and a targeted therapy? Can three be better than two? So there are trials both in the front-line setting and in the refractory setting, looking at these novel therapies in the one bucket. And then there are also trials looking at these combinations and different ways of mixing and matching therapies that we already have to optimize patient outcomes?

Lisa Hatfield:

Great. Thank you for that description of clinical trials, too. That’s very helpful.

Okay. Now it’s that time where we answer questions that we’ve received from you. Please remember that this is not a substitute for your medical care. Always consult with your medical team. So, Dr. Ornstein, how do you explain a kidney cancer diagnosis to your newly diagnosed patients, and what are the priorities for those newly diagnosed patients?

Dr. Moshe Ornstein:

When I’m looking at a patient and their family with a newly diagnosed kidney cancer, I’m trying to think to myself a couple of things. Number one, how can I make it as easy to understand without withholding any information? How can I be as encouraging as possible, but at the same time without misleading the patient in terms of what’s to come? The way I break it down is into two main categories. There is the patient that presents with a localized kidney cancer.

So they came to the emergency room because they were having belly pain, and they were found to have a big mass growing in their kidney that is proven to be kidney cancer. And then there’s the patient who has advanced disease, metastatic disease that has spread beyond the kidney. Either they came in with metastatic disease, in other words, their kidneys in place, and they have cancer beyond the kidney. Or they already had a surgery a year or two ago, and now they come back, and the cancer has returned elsewhere in the body.

So for the patient that comes with a localized kidney cancer or kidney cancer limited to the kidney, I talk to them about what the diagnosis means in terms of what subtype of kidney cancer is it, meaning although most kidney cancers are clear cell renal cell carcinoma, there are other types of kidney cancer. And I want them to have a good handle in terms of what they have, both so that they know and they have all the information they need, but also because I understand that most patients, or at least many patients are going to look for more information elsewhere.  And without understanding the histology, the type of kidney cancer they actually have, I worry that they’re not going to get the right information. So I try to be very clear about what stage is it based on the scans or if they’re coming to see me after their surgery, what stage is it after the surgery?

What does it mean when something is a grade 1 versus a grade 2 versus a grade 3 in terms of what the cells look like under the microscope? That’s about the cancer. And then I talk to them about what we’re going to look for in the future. So again, we’re talking now about the patient with localized kidney cancer. I try to go through with them what the risk of that localized cancer is in terms of what the odds are of it coming back. We talk about what kind of surveillance, what kind of watching or monitoring of the cancer are we going to do, how often they’re going to get CAT scans.

So really try to give them the big picture about what cancer they have, what the outlook is, and what we’re going to do to keep a close eye on them. For the patient who has an advanced cancer, in some ways it’s similar. When I say advanced, I mean a cancer that has spread beyond the kidney that’s going to require therapy, immunotherapy, targeted therapy, a clinical trial, whatever it might be.

And again, super important for them to understand, is this a clear cell kidney cancer? Which is the most common? Is it a papillary kidney cancer? Is it something else? Then we talk about what the different treatment options are. What does the short term look like in terms of side effects? What does the short term look like in terms of getting the cancer under control? What does the long-term outlook look like? What are the possible long-term side effects? And then what are we going to do to monitor? Are we doing CAT scans? Are we doing MRIs? Are we doing imaging of their brain?

So again, first and foremost, what’s the nature of the diagnosis, what the treatment options are and likely side effects, what they need to look out for, and then how we as a medical team are going to monitor this over hopefully the long run.

Lisa Hatfield:

We have a patient asking if you can speak to a typical patient history associated with kidney cancer and is there a common factor, or I think that they’re asking is there a cause that you see frequently for kidney cancer?

Dr. Moshe Ornstein: 

Yeah, this is such a common question, Lisa, because patients want to know I have this cancer, what caused it? And generally, we just don’t know the answer to that. And I tell that to patients, it’s generally not something that somebody did that caused this kidney cancer. We do have known risk factors for kidney cancer, whether it’s obesity, smoking, high blood pressure, chronic kidney disease. So there are certain risk factors and associations, but it’s really difficult for a specific patient to be able to pinpoint this caused the kidney cancer. And I think it’s reassuring for patients to know that as a general rule, it’s not something that a patient did that caused the kidney cancer, and it’s not somebody’s fault that they have the kidney cancer.

Lisa Hatfield:

Okay. Thank you for that. What is hereditary renal cell carcinoma, and can you speak to the instance that you’ve seen in your practice? And third part of that question is, how can I protect my family?

Dr. Moshe Ornstein: 

It’s a loaded question, Lisa. And the truth is, you know, patients come in and many patients are not concerned about their well-being, they’re more concerned about their family, and they want to know, “Is this something that’s going to impact my children? Do my children need to be screened for kidney cancer at an earlier age or screened at all?” Because generally we don’t screen people for kidney cancer. 90+, maybe even as high as 95 percent of kidney cancer is what’s called sporadic.

In other words, it just comes out of the blue. I tell patients in some ways it’s just bad luck. It’s not anything they did. It’s not something that they got a gene from their mother or father that caused it, and it’s not something that they’re going to pass down to their children. There’s a very small percentage, maybe about 5 percent or so of kidney cancer that’s hereditary, that does have, you know, a genetic association. That is something that they can potentially pass down, and they may have received that gene from a parent.

It’s exceedingly rare. We think about VHL syndrome, we think about something called hereditary papillary, tuberous sclerosis complex, Birt-Hogg-Dubé, but the overwhelming majority are sporadic, not associated with any specific gene in terms of a gene passed down from parent to child. What I would say is when I start to think about an inherited kidney cancer, I’m thinking more about a very young patient who comes in with kidney cancer, where we don’t expect young patients generally to have kidney cancer, or a patient who shows up with kidney cancer that’s in both of the kidneys. So there are certain unusual features that would lead a doctor to think about a hereditary or genetically associated kidney cancer. But overwhelmingly, it’s sporadic. Children are not necessarily at a higher risk, don’t need to be screened. But for these small features we do in clinic, keep an eye out for that.

Lisa Hatfield:

Okay. Thank you for that. So when you have a patient who comes in with those more unusual presentations, do you recommend that they get some type of genetic testing done, so they can be aware for their family members that maybe they should be screened?

Dr. Moshe Ornstein:

Yeah, absolutely. I mean, if there’s an unusual feature, either a feature associated with tuberous sclerosis complex or something called Birt-Hogg-Dubé, or a young patient with advanced kidney cancer where we don’t expect it, or a patient that shows up with cancer in both of their kidneys and nowhere else, that will trigger us to send the patient to a genetic counselor to do a more thorough family history and talk about what they might be looking for in terms of genetic testing.

Lisa Hatfield:

Okay. Thank you. All right. Another person watching is asking, are there known occupational exposure risk factors for kidney cancer?

Dr. Moshe Ornstein:

This is a great question. You know, we know that with certain cancers, there are classic occupational exposure risks. People want to know, “If I worked in a coal mine, am I more likely to get this kind of kidney cancer? What if I’m a Vietnam veteran and I was exposed to Agent Orange, is this more likely?” Really difficult to find those associations.

I would say that probably the biggest ones are going to be, again, smoking, which I don’t know is so much an occupational hazard, although secondhand smoke is a real risk factor for cancers. Asbestos. So people who worked around a lot of asbestos, that can be a risk factor even for kidney cancer. I know we usually think about it as lung cancer mesothelioma, but definitely for kidney cancer as well in some studies. And then certain forms of gasoline exposure. I will tell you that I’ve taken care of a lot of patients and a lot of people who have kidney cancer and have never been able to isolate an occupational exposure. But looking in the literature, we’re really looking more for asbestos, certain gasoline, secondhand smoke, things like that.

Lisa Hatifeld:

Okay. Thank you. Another patient is asking if there are any specific diets or diet recommendations for kidney cancer patients, especially as they relate to managing side effects of the treatment.

Dr. Moshe Ornstein:

We get asked all the time. And my general answer that I give, and again it’s sort of tongue in cheek, is I tell the patients the same diet that I should be following is the diet that I would recommend to you. I tell patients it’s a well-balanced diet, the right amount of carbs, the right amount of protein, the right amount of Snicker bars. I’m not a get rid of all your sugar or don’t drink any caffeine person. So I think in terms of sort of being data-driven, I would say a generally well-balanced diet.

However, some of these therapies, particularly the TKIs can cause diarrhea. They can cause mouth sores, they might change how you feel. So even though a well-balanced diet is great, we also encourage patients and empower them and their families to follow a diet that’s going to sit well with them given the therapy that they’re on.

So I care about diet much less in the sense of how’s the diet going to affect the cancer and the long-term outcome, and much more in eat the foods that your body will accept. If your body is tolerating more pasta now than it did before, because that helps your gut and therefore you’re able to stay on therapy, wonderful. If you’ve become more of a protein person because that doesn’t instigate the diarrhea, also great. If you need food that has more salt or less spice because of how your mouth feels because of the therapy, then that’s what you need to eat. So really to pay attention to their own bodies and know that whatever they fall on in terms of their diet, again, taking the extremes out of the equation, it’s okay.

Lisa Hatifeld:

You’ve made a lot of patients happy with that response. Thank you.

Dr. Moshe Ornstein:

I think I may have made a lot of spouses unhappy. But again, I think it’s important to work as a team with the medical team and with family.

Lisa Hatifeld:

Great. Thank you. One last question. A patient is asking, “I’m newly diagnosed and my stage of kidney cancer is unclear as I wait for further review. What questions should I be asking my team? It’s very scary to know you have cancer, but unclear of how serious.”

Dr. Moshe Ornstein:

The first thing I would tell this patient is, it’s okay to be scared. And there’s no right or wrong way to feel while you’re waiting for the uncertainty to be settled. Some people have trouble sleeping, some people cry, some people shy away from interacting with their friends and loved ones. And there’s no right and wrong way to respond.

Once you’re comfortable, once a patient is comfortable saying, however I feel emotionally is okay, now we can talk about what you should be asking and what they should be asking. I always try to frame it as what am I looking for in the short term, and what am I looking for in the long term? And I think it’s important to ask the team, ‘Is this cancer something that we’re going to treat with a goal of eliminating it, or is this cancer something that I’m more likely to be on therapy for the long term?”

Again, a short-term question and a long-term question, and the team can give a general overview. I think it’s okay for a doctor to say, I don’t know, or I don’t know yet. I think if you have a doctor that says that, you’re probably very lucky because they’re comfortable being honest and telling you what they know and what they don’t know and what they’re going to do to get the information.

So I would say the questions to ask are, “What’s the next step in the investigations, whether it’s additional scans or additional biopsies? What are the chances that this is something that’s only going to be a short-term issue, and what are the chances that this is something that I’m looking at sort of a chronic condition for the long term?”

Lisa Hatifeld:  

Great. Thank you so much for that answer and showing compassion when you answered that question too. That’s a difficult diagnosis to receive. So thank you for that.

Dr. Ornstein, thank you so much for joining us today. We really appreciate your time and expertise. On behalf of all patients, including myself, a cancer patient, we’re so thankful for the opportunity to listen to your answers, for you to let us ask questions. So we appreciate your time. Thank you so much.

Dr. Moshe Ornstein:

Yep. Really my pleasure to be here and thanks so much for the opportunity.

Head and Neck Cancer Clinical Trials | What Are the Benefits?

Head and Neck Cancer Clinical Trials | What Are the Benefits? from Patient Empowerment Network on Vimeo.

What benefits can head and neck cancer clinical trials bring to patients? Expert Dr. Ezra Cohen discusses clinical trial participation rates, explains how trials move research forward, and details questions to ask your doctor when considering joining a clinical trial. 

Dr. Ezra Cohen is a medical oncologist, head and neck cancer researcher and Chief Medical Officer of Oncology at Tempus Labs.

Download Resource Guide

See More from Evolve Head & Neck Cancer

Related Resources:

Head and Neck Cancer Treatment and Research Updates

Head and Neck Cancer Treatment and Research Updates

Head and Neck Cancer Research | How Innovation Leads to Advances in Care

Head and Neck Cancer Research | How Innovation Leads to Advances in Care

How Is Head & Neck Cancer Treatment Approached?

How Is Head & Neck Cancer Treatment Approached?

Transcript:

 Katherine:

A key part of research moving forward is the clinical trial process. Can you talk about the benefits of patient participation?  

Dr. Cohen:

Of course. And I’m certainly a firm believer. It’s been part of what I’ve been doing for, again, over 20 years. And, so, certainly I think clinical trials are a critical component of our ability to produce better outcomes and move forward. The sobering reality is that only 3  percent of adult patients actually participate in clinical trials.  

Katherine:

Wow. 

Dr. Cohen:

And if we’re really going to move the field forward and be where we want to be with cancer, and that is to either cure as many people as we can or turn cancer into a chronic disease, better yet in many individuals, detect it early and prevent it. We need much greater participation in clinical trials. So, that being said, nothing is going to happen, we are not going to make strides forward in therapeutics and diagnostics, whatever in prevention, without clinical trials. Even the basic standards of care that we engage in now, and I mentioned that drug pembrolizumab (Keytruda), that was approved on the basis of a large randomized clinical trial.  

Now, having said that, I think patients – part of the responsibility is on patients to ask their provider, “Well, what about clinical trials?” There is almost always a clinical trial that is available given a specific setting. The challenge is that it may not be available where the patient is seeking care. And there is a large disconnect in that regard. Now, there are different organizations that are trying to improve that, improve the diversity of participation in clinical trials and the equality of availability of clinical trials to the entire population, not just people going to large academic medical centers. But, that’s the first thing I would tell patients, is part of the list of questions that you have for your provider, especially at the beginning and throughout the entire process, is there a clinical trial that would be applicable to my situation?  

And if the answer is, “Well, no.” The follow-up should be, “Well, what about in the area?” Or, “What about something that may not necessarily be available in this practice but what about somewhere else in town or somewhere I could access?” Of course, that may not be possible for all patients. But if it is, I would encourage all people to ask that question. And then there is a large responsibility, I think, on providers to make clinical trials available if we’re going to improve that number from 3 percent. 

There are a lot of exciting agents being studied now in head and neck cancer – immunotherapy drugs, antibody drug conjugates, novel mechanisms, cell therapies, vaccines. So, there’s certainly no paucity of interest in developing drugs in head and neck cancer and technologies as well. So, there are clinical trials available, and I think we, as a community, need to make a – need to do better in enrolling patients on those studies. 

Self-Advocacy in Myeloma Care | Advice From an Expert

Self-Advocacy in Myeloma Care | Advice From an Expert from Patient Empowerment Network on Vimeo.

What questions should myeloma patients ask their doctor about a treatment plan? Myeloma expert Dr. Omar Nadeem shares key advice and emphasizes the importance of patients engaging with their healthcare team in treatment decisions.

Dr. Omar Nadeem is the Clinical Director of the Myeloma Immune Effector Cell Therapy Program and Associate Director of the Multiple Myeloma Clinical Research Program at the Dana-Farber Cancer Institute. Learn more about Dr. Nadeem.

Download Resource Guide

See More from Evolve Myeloma

Related Resources:

Clinical Trials for Myeloma Treatment | Essential Information for Patients

Clinical Trials for Myeloma Treatment | Essential Information for Patients

What Should Myeloma Patients Ask About Developing Research

What Should Myeloma Patients Ask About Developing Research?

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Transcript:

Katherine:

What questions should patients be asking their provider about a proposed treatment plan?  

Dr. Nadeem:

Yeah. I think because myeloma therapy’s so nuanced and much of this is still in clinical trials or under investigation about what to do with some of these results, I would say, as a whole, it’s important to know which tests the physician looks at to determine how you’re doing, and kind of what their assessment of that result is. So, for example, if somebody’s had a 50 percent reduction in the amount of abnormal protein in the blood, is that sufficient, or should we be aiming for a number that’s much higher than that? 

Some of that depends on kind of where they are in their treatment course, but that’s a very sort of reasonable question to ask your physician is that where do you see my response now, let’s say six months into therapy, and is this adequate, and what is now, after we have all this information, what is my roadmap going forward to try to keep this disease in check? 

Katherine:

Yeah. Well, that’s great advice, Dr. Nadeem. Thank you. I’d like to turn to self-advocacy, Dr. Nadeem. Why is it so important that patients engage in their care treatment decisions?  

Dr. Nadeem:

Yeah. As I mentioned, myeloma therapy is so individualized now and we can sit here, look at the trial data, get very into the weeds and technical about this therapy with this approach as X or Y higher response rate.  

Or MRD-negative rate, but in reality, we’re dealing with people and we’re dealing with people that have lives.  

They have all their priorities, and until you share that with us, it’s very difficult for us to know exactly what’s important to you. So, what I may consider to be kind of the “best therapy” for you may not make sense for you because of all the priorities that you may have, and I think it’s so important to advocate for yourself and not be afraid to bring that up to your physician because I think many patients kind of hold that stuff in for a long time because they don’t want it to impact their care. But I would argue the other way around.  

Tell us. Tell us exactly what you prioritize. Tell us if you can’t be out of commission for work for X amount of time because of a stem cell transplant. We now have options. We now have options for patients because of all these amazing new therapies for myeloma and we can come up with a very individualized treatment plan for you based on your priorities.  

Katherine:

If a patient is feeling like they’re not getting the best care or they’re uncomfortable with the care they’re receiving, what steps should they take to change that?  

Dr. Nadeem:

Yeah, I think that’s very difficult because this is a complex system. Medical systems are getting even more and more complex. They’re busy. Everybody’s busy: busy offices, labs, radiology. We’re all feeling that. It doesn’t matter where you are. So, I think it’s important to raise those concerns, number one, to your practice that you’re being seen at because they would like to see that feedback, right? So, kind of see what is something that they can perhaps improve upon. I think it’s always important, like we just said, to advocate for yourself and raise some of these issues and not be afraid of that.  

We’re all in this together, right, so I think ultimately, we’re all trying to take the best care of you and we would need to know which part of that may or may not be working so well.   

Evolving Myeloma Treatment Options | CAR T-Cell Therapy

Evolving Myeloma Treatment Options | CAR T-Cell Therapy from Patient Empowerment Network on Vimeo.

What is CAR T-cell therapy, and who is it right for? Dr. Omar Nadeem of Dana-Farber Cancer Institute discusses the role of this therapy in myeloma care and shares an update in ongoing CAR T-cell therapy clinical trial research.

Dr. Omar Nadeem is the Clinical Director of the Myeloma Immune Effector Cell Therapy Program and Associate Director of the Multiple Myeloma Clinical Research Program at the Dana-Farber Cancer Institute. Learn more about Dr. Nadeem.

Download Resource Guide

See More from Evolve Myeloma

Related Resources:

Clinical Trials for Myeloma Treatment | Essential Information for Patients

Clinical Trials for Myeloma Treatment | Essential Information for Patients

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Personalized Medicine for Myeloma Treatment | What Patients Should Know

What Should Myeloma Patients Ask About Developing Research

What Should Myeloma Patients Ask About Developing Research?

Transcript:

Katherine:

Well, I’d like to talk about some new and emerging therapies in myeloma, starting with CAR T-cell therapy. Can you talk about who this treatment option might be appropriate for?  

Dr. Nadeem:

So, yeah, just to kind of give folks background, CAR T-cell therapy is a form of immunotherapy, where we take out an individual’s T-cells and then re-program them, essentially, to recognize myeloma cells. Right now there’s two approved CAR-T products for multiple myeloma, both in the relapse refractory setting. It’s really for patients that have had four or more lines of therapy.  

So, that’s a lot of different combinations that we currently have available. Those therapies stop working before patients are actually eligible for CAR-T cells at the moment. Both of these CAR T-cell products have been gamechangers in terms of improving prognosis for patients.  

The good thing about CAR-T cells is that it is a one-and-done treatment. So, patients, when they go through that initial phase of therapy, they are then off therapy, although we are now starting to study certain therapies that we may administer after CAR-T cells to get them to last even longer than they currently do, but that’s still in, for example, that’s one of the clinical trials or many of the clinical trials that are currently ongoing now, to try to answer that question.  

So, a lot of patients can be eligible for CAR-T cells. They have to have the prerequisite amount of therapies. Again, there are some sort of baseline fitness characteristics that we look at for patient’s ability to tolerate it. But as a whole, I consider CAR T-cell therapy more broadly applicable to myeloma patients than compared to, let’s say, a stem cell transplant.  

Katherine:

How has this therapy revolutionized myeloma care? 

Dr. Nadeem:

Yeah, before the first approval, now a few years ago, in this space we didn’t really have anything like this to offer patients. So, many of the combinations and other compounds that were in clinical trials would have a response rate somewhere around, let’s say, 30 percent. So, 30 percent of patients may respond to that therapy in that space, and that may only last a few months, and that was considered successful not that long ago. Now, with CAR T-cell therapy and bispecific antibodies, these therapies are highly efficacious.  

You see response rates of 70 to 100 percent in some of these immunotherapies, and what that’s translating into is patient’s disease staying away for a year or two years, even three years in some of these clinical trials. And again, this is completely unprecedented compared to what we had before.  

Katherine:

I understand that there are a number of clinical trials for different types of CAR T, or even using it earlier in the disease. Can you share updates in CAR T-cell therapy research? 

Dr. Nadeem:

Yeah, so, exactly as you pointed out, there have been trials already, actually, that have been completed, Phase III studies looking at CAR T-cell therapies in earlier relapses.  So, patients that have had either one of two lines of therapy. 

Both our CAR-T therapies have been compared to standard of care in that space and have shown superiority, and this is something that we all have been kind of waiting for to see if you deploy it earlier, perhaps you’re going to see even greater benefit, and that seems to be the case in some of these trials, and now we’re awaiting, hopefully, approval of some of these CAR T-cell therapies to be administered earlier because in fifth line, it’s very different than treating patients in second or third line, which I think will really vastly improve our ability to deliver this therapy to many patients, as it can be quite challenging for patients that are in fifth line, to allow them to go through the process of CAR-T cells and then having them be administered.  

I was looking at it head-to-head with stem cell transplant, as I mentioned before, and this is in the context of quadruplet and induction therapy followed by either CAR-T cells or stem cell transplant, and then followed by maintenance therapy. So, really trying to see if I can overcome what we typically have achieved with stem cell transplantation.  

We also are doing some studies even before that. So, patients, again, in high-risk smoldering myeloma, which we know have an increased risk of developing newly diagnosed disease in the next few years, perhaps that could be the time where we can give some of these immunotherapies, and that’s some work that we have going on at our center. 

Clinical Trials for Myeloma Treatment | Essential Information for Patients

Clinical Trials for Myeloma Treatment | Essential Information for Patients from Patient Empowerment Network on Vimeo.

How do clinical trials advance treatment options for myeloma? Dr. Omar Nadeem discusses the important role of clinical trials in improving patient care, key questions to ask your care team about trial participation, and the benefits of seeing a myeloma specialist. 

Dr. Omar Nadeem is the Clinical Director of the Myeloma Immune Effector Cell Therapy Program and Associate Director of the Multiple Myeloma Clinical Research Program at the Dana-Farber Cancer Institute. Learn more about Dr. Nadeem.

Download Resource Guide

See More from Evolve Myeloma

Related Resources:

Next Generation Myeloma Treatment Options

Next Generation Myeloma Treatment Options

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Personalized Medicine for Myeloma Treatment | What Patients Should Know

Myeloma Research Highlights From ASH 2023

Myeloma Research Highlights From ASH 2023 

Transcript:

Katherine:

I’d like to start with the importance of a patient’s healthcare team. What are the benefits to seeking care with a myeloma specialist, even if it’s just for a second opinion or a consult? 

Dr. Nadeem:

Yeah, so, myeloma is a little less than 2 percent of all cancers, and it’s the second most common blood cancer, so certainly not rare. With that being said, if you go to a general community practice, they don’t typically see too, too many patients with this disease. So, alongside that, we have so many different treatment options and combinations and these, as I mentioned, immune therapies.  

And other therapies that are only actually carried out at academic centers for now, such as stem cell transplants, and CAR T-cell therapy.  

I think it’s important to kind of meet with an academic provider just to get a sense of what the patient may be facing, both in that immediate time, but also in the future, because a lot of myeloma therapy is lifelong. And in that case, you do have to come up with a plan for your whole treatment in a way early. So, it’s important to kind of one: hear it from another person, and then two: really sort of figure out what the outlook would look like for the individual patient.  

With that being said, many of our myeloma regimens that are approved can very easily be given at the local provider, and that’s usually our preference, for patients to be treated closer to home. So, ultimately, this is another way for patients to get input about their treatment program, but also talk about the future.  

Katherine:

That makes sense. Specialists at academic medical centers are typically more involved in research and clinical trials. 

And patient participation is essential to advancing medicine. So, how do clinical trials impact myeloma care? 

Dr. Nadeem:

Well, everything that we have available today for myeloma therapy was once in a clinical trial. So, all these promising therapies usually start in early phase studies and move on to Phase II and Phase III studies, and then those are the ones that the FDA uses to approve a particular combination.  

So, it all depends on kind of where someone is in their disease course. It also kind of depends on what their preferences may be in terms of taking on something that is beyond standard of care. So, as part of any clinical trial in whatever phase it may be, whether its newly diagnosed multiple myeloma, even smoldering myeloma, which is one step before that, relapsed/refractory myeloma…  

At each step of the way, there are clinical trials that are there trying to improve upon what’s already out here, right? So, we are, despite all these amazing advances, unfortunately, the disease is still not curable for a vast majority of patients.  

In that case, how do we move to that cure, or how do we kind of advance the disease even beyond this? And a clinical trial is a way to do that.  

Katherine:

What type of patient is most appropriate for a clinical trial? 

Dr. Nadeem:

So, there are criteria that each clinical trial uses in terms of eligibility. Some of that has to do with the disease characteristic itself, kind of where somebody is in their disease course, but many times it’s also patients’ fitness, organ status in terms of kidney function, their blood count to some extent, heart function, etcetera. There are some sort of minimal prerequisite guidelines that we have to enroll patients in trials. So, it really, again, depends on where somebody is in their disease course and what they may be willing to take on beyond what may be offered to them as part of standard of care.  

Katherine:

What questions should patients be asking if they’re entrusted in participating in a clinical trial? 

Dr. Nadeem:

I think the important thing is to sort of first recognize what’s available to them as part of standard of care and then what the clinical trial is trying to answer.  

So, for example, if it’s newly diagnosed multiple myeloma, we now have quadruplet regimens that we give to patients at the time of their diagnosis, and then the next natural question for eligible patients that now comes up is whether they should do a stem cell transplant or not.  

And alongside that goes with all these advances in immune therapies, such as CAR T-cell therapies and bispecific antibodies. And there are now trials looking at those therapies and comparing them, for example, to stem cell transplant to try to answer the question “Can we get even beyond something like a stem cell transplant?”  

So, that’s one example of a trial where a patient may be interested in saying “Okay, well, a transplant may be my standard path, but what if I try to enroll in this study and get randomized, for example, to the CAR-T arm? Then, perhaps, I’m getting access to some of these therapies early and maybe that’s going to improve my outcomes.” 

Thriving With Small Cell Lung Cancer Resource Guide

Download Resource Guide

PEN-202_SCLCThrive_Guide

Download Resource Guide

See More from Thrive Small Cell Lung Cancer

Expert Advice for Patients With Small Cell Lung Cancer

Expert Advice for Patients With Small Cell Lung Cancer from Patient Empowerment Network on Vimeo.

Dr. Triparna Sen, a leading small cell lung cancer (SCLC) researcher and expert, shares key advice for patients. Dr. Sen stresses the importance of working closely with your doctor, asking about clinical trials options, and the benefits of support groups.

Dr. Triparna Sen is an associate professor in the department of oncological sciences and co-director of the Lung Cancer PDX Platform at the Icahn School of Medicine at Mount Sinai in New York. Learn more about Dr. Sen.

See More from Thrive Small Cell Lung Cancer

Related Resources:

Essential Small Cell Lung Cancer Testing

Essential Small Cell Lung Cancer Testing 

Advances in Small Cell Lung Cancer Research | Hope for the Future

Advances in Small Cell Lung Cancer Research | Hope for the Future

Understanding Small Cell Lung Cancer Treatment Options

Understanding Small Cell Lung Cancer Treatment Options

Transcript:

Katherine:

What three key pieces of advice would you have for a patient who has just been diagnosed with small cell lung cancer?  

Dr. Sen:

First of all, I would like to say definitely, definitely work with your physician. They are trying to do their best for you. Work with your physician. Follow the treatment regimen that they give. Ask about clinical trials that you are eligible for and that you can enroll into. Then, of course, having a support group. So, there are many patient advocacy groups right now for non-small cell and small cell lung cancer.  

I think being a part of such a patient advocacy group where you have people going through the same journey, I think, it really helps. It helps you sort of manage your disease better. It helps you stay hopeful when you hear about other people’s sort of prognosis and if they have durable benefits from drugs. So, I think having a support group is very important. If there is an ability for you to contribute to research in terms of giving blood or tissues, if your physician is saying that you could be eligible for that, I think a contribution to research is really key. 

Because looking at the disease mechanisms in the clinical tissue is sort of where for us it is absolutely golden. We go there. We look at the disease mechanisms and tissues. If there is an opportunity for you, then I think it should definitely be explored.  

Finally, I would like to say we are really trying as researchers to really understand the disease better. We’re trying to do that better. I hope and I pray that we go faster with it. But I think there is hope right now for patients with small cell lung cancer. The research is really progressing better. There are many clinical trials.  

So, I think stay hopeful and have a peer support group who can take you through this quite difficult journey.  

Katherine:

Why should patients consider consulting with a lung cancer specialist?  

Dr. Sen:

I think it’s crucial because these lung cancer specialists really know the current state of the art treatments. They are thought leaders. They participate in trials. They actually sit on advisory boards with companies.  

They are strategizing the entire treatment landscape for this disease. So, if you go to a lung cancer specialist, you’re more likely to get the most updated knowledge about what treatments are out there, what you qualify for, what are the clinical trials out there, and what are working in patients. This is not just for small cell. There are many, many trials that are happening in non-small cell also. So, whatever your diagnosis is, a specialist should be able to tell you what your options are. You really want to know about your options. Your options about biomarker testing.   

Your options about screening. Your options about trials. I think a lung cancer specialist can really guide you towards that.   

Katherine:

Dr. Sen, thank you so much for joining us today. It’s been a pleasure speaking with you.  

Dr. Sen:

Thank you.  

Advances in Small Cell Lung Cancer Research | Hope for the Future

Advances in Small Cell Lung Cancer Research | Hope for the Future from Patient Empowerment Network on Vimeo.

What new treatments are being studied for small cell lung cancer (SCLC)? Dr. Triparna Sen, a leading researcher in the field, shares promising updates, including advances being made with LSD1 inhibitors, DDR (DNA Damage Response) inhibitors, and DLL-3 targeted therapies.

Dr. Triparna Sen is an associate professor in the department of oncological sciences and co-director of the Lung Cancer PDX Platform at the Icahn School of Medicine at Mount Sinai in New York. Learn more about Dr. Sen.

See More from Thrive Small Cell Lung Cancer

Related Resources:

Essential Small Cell Lung Cancer Testing

Essential Small Cell Lung Cancer Testing 

Expert Advice for Patients With Small Cell Lung Cancer

Expert Advice for Patients With Small Cell Lung Cancer

Understanding Small Cell Lung Cancer Treatment Options

Understanding Small Cell Lung Cancer Treatment Options

Transcript:

Katherine:

Dr. Sen, you are a leading researcher in the field. What is the latest research news that you can share with us about small cell lung cancer?

Dr. Sen:

There’s a lot of great research going on in my lab and labs all across the world. I think for the first time in a very long time, we are really trying to dissect the biology of small cell.   

It has been a research in making for many years. I think we have now really come to a point where we are really trying to understand the disease. I’ll go into a little more about the questions you are trying to answer. So, one of the main questions or one  of the main things that kind of is a hurdle to getting durable treatment options is that the frontline chemotherapy and immunotherapy doesn’t work as well as they should even for the approved regimens, which is the chemotherapy and the immunotherapy.  

The patients often do not have durable benefits. Even if patients have durable benefits, it’s only in a very minority of patient population which means in only about 10 to 15 percent of the total patient population actually do have any benefit from the frontline treatment. So, the main question that we are trying to answer is that why do these patients not respond to immunotherapy and chemotherapy in the frontline.  

What are the mechanisms of resistance to chemotherapy and immunotherapy? Primary resistance, what I mean by primary resistance is that patients who never respond. The disease comes back even while they’re getting the frontline chemo. So, the primary resistance, the mechanisms. Of course, when they have acquired resistance after the maintenance regimen when they come back, why are these patients having this acquired resistance to chemotherapy and immunotherapy? Because only when we understand resistance mechanisms will we be able to then come to the combination strategies.

That’s the next area of research is that once we understand the mechanism of chemotherapy and immunotherapy resistance is then coming up with effective combination therapy. So, what should we combine with immunotherapy in order to make immunotherapy better? I’ll give you an example from the research that we did. 

So, our lab focus is, as I said, on making immunotherapy better. What we understood is that there are certain epigenetic modifiers like LSD1.  

Repressing these, repressing LSD1, with a small molecule inhibitor actually augments or benefits the response to immunotherapy. So now, we are looking at LSD1 inhibitors in combination with immunotherapy. That’s one area that we are focusing on. The second are that we published extensively on is DNA damage response inhibitors which really works in combination with immunotherapy and makes immunotherapy response better.  

Now, we are investigating that in the lab the combination strategies of combining these DNA damage response inhibitors with immunotherapy. So, combination strategies. I think always coming up with novel targets. I will mention there are many novel targets that are right now in the clinical trials actually showing really, really encouraging data.  

I’m talking about DLL3 targeted BiTEs or ADCs we have seen that are showing preliminary data. We have seen a really good really good response in patients. So, finding these targets that are very specific for small cell and that can work in these unique population of patients.  

So, DLL3 targeted agents. There are agents that target B7-H3. So, we are looking at these novel targets and where they could fit in the current therapeutic regimen. Finally, since small cell lung cancer is not a surgical disease, we have to look for other options to find biomarkers. So, liquid biopsy. Liquid biopsy, what I mean by that is understanding the disease not just from tissue but also from blood.  

There’s a lot of research that’s happening in understanding the biology of small cell from blood draws from these patients.  

So, the field of using liquid biopsy or understanding the disease from blood draws is one of the areas that many labs, including ours, are focusing on, and how we can utilize these blood samples to then monitor the disease and also understand the resistance mechanisms to various drugs. I think these are the areas that we are investigating and seems, to me, very important areas that we need to address in order to really manage small cell lung cancer.   

Katherine:

What do these advances mean for small cell lung cancer patients? Are you hopeful?  

Dr. Sen:

Oh, yes. Of course. We’re always hopeful. That’s the goal, right. The goal is to have effective therapies that work and that works for a long time. That also benefits the patients in terms of quality of life which means without very severe adverse effects.   

So, very hopeful. Because I think what was limiting us for all those years for the last 40 to 50 years is that we really did not understand the complexity of small cell lung cancer. It is a very complex disease. It is very different from non-small cell lung cancer which has these mutations that you can target drugs against. So, there are this EGFR mutations and KRAS mutations in non-small cell.  

But small cell, it’s not that. It is not a disease where we have these GATA function mutations that we can devise therapies against. It’s a very different disease. The disease is aggressive. The disease progresses fast, and it also changes its physiology very fast. So, I think for the first time, we really are trying to understand the biology. What that helps is then to come with very informed decisions about therapy.  

So, yeah, I’m very hopeful. Because I think we have now targets that we are actually seeing benefits in patients. I think the more and more we understand resistance mechanisms, we’ll also be able to manage that better.   

Katherine:

That’s very promising news. 

Understanding Small Cell Lung Cancer Treatment Options

Understanding Small Cell Lung Cancer Treatment Options from Patient Empowerment Network on Vimeo.

How is small cell lung cancer (SCLC) treated? Dr. Triparna Sen discusses treatment options for patients with small cell lung cancer, both first-line and second-line therapies, and the important role of clinical trials in patient care. 

Dr. Triparna Sen is an associate professor in the department of oncological sciences and co-director of the Lung Cancer PDX Platform at the Icahn School of Medicine at Mount Sinai in New York. Learn more about Dr. Sen.

See More from Thrive Small Cell Lung Cancer

Related Resources:

Essential Small Cell Lung Cancer Testing

Essential Small Cell Lung Cancer Testing 

Expert Advice for Patients With Small Cell Lung Cancer

Expert Advice for Patients With Small Cell Lung Cancer

Advances in Small Cell Lung Cancer Research | Hope for the Future

Advances in Small Cell Lung Cancer Research | Hope for the Future

Transcript:

Katherine:

How do test results impact care? 

Dr. Sen:

So, you know, once the doctor has confirmed the small cell lung cancer and we have confirmed what stage it is at – what I mean by staging is that it could be either a limited stage disease which is an early stage small cell, or it could be an extensive stage of small cell. The treatment for those two are quite different. So, if it is an early stage or limited stage, patients are usually treated with chemoradiation. If it is an extensive stage or a metastatic small cell, then patients are usually given a standard of care which is chemotherapy in culmination with immunotherapy which is an antibody against PD-L1.  

Katherine:

You’re talking about treatment options that are currently available for small cell lung cancer. What about targeted therapies?  

Dr. Sen:

There aren’t very many therapeutic strategies that are targeted therapies as we speak like we hear from non-small cell lung cancer.  

So currently, like I mentioned, the frontline treatment for small cell lung cancer is with chemotherapy and immunotherapy and maintenance with immunotherapy alone.  

Once the patient relapses, which often is the case – all patients actually have resistance to the frontline chemo-io (chemoimmunotherapy) at some point in time. Once they have a relapse disease, the second line of therapy until now is with either topotecan or irinotecan which are two topoisomerase inhibitors or with lurbinectedin which is in the second line.  

So, when it comes to targeted therapies, so far we a have seen, you know, the conventional way that we think about EGFR inhibitors or KRAS inhibitors, it hasn’t been the case so far with small cell lung cancer. It’s very limited in the current approved setting. But there are many clinical trials that are investigating several targeted therapies that are either targeting – I can speak about that more as I talk about research strategy. But there are many targeted agents that are targeting surface targets like DLL3, B7-H3, or SEZ6. There are other targets that are targeting things like DNA damage repair, proteins, or epigenetic regulators like LSD1. But so far in the approved setting, it is quite limited.  

Katherine:

When we look at what therapies are available, what treatment options are available, what are some typical side effects? How are they managed?  

Dr. Sen:

Some of the major side effects that you see, especially with a frontline chemo-io (chemoimmunotherapy), are very common like you see with other cancer types. Also, it’s usually myelosuppression.  

I think it is prevented or is managed either by dose reduction or treatment delays or treated with transfusion. There has been research that CDK4/6 inhibitors, trilaciclib, when treated with in combination with chemotherapy can bring down the side effects that we see with chemotherapy.  

Some of the immunotherapy related adverse events includes pneumonitis, colitis. They are usually treated with early steroids, treatment withholding, and also it could be leading to permanent discontinuation of the treatment if the adverse events are really severe. Those are mainly what we see we the chemo-io (chemoimmunotherapy) regimen that is given up front.  

Katherine:

Okay. What questions should someone be asking about their proposed treatment plan?  

Dr. Sen:

Right. So, I think, of course, first is what stage. The treatment will depend upon the stage of small cell. Usually, on the frontline, everyone is given chemotherapy and immunotherapy. 

It’s a systemic therapy that’s being given. But I think the patient should be asking questions like are there clinical trials available for me. Because there are multiple clinical trials right now in the frontline and the second line setting.  

So, I think definitely the patient should ask about the clinical trials that the qualify for. In terms of contributing to research, I think if there are options for them to either sign up for blood collection protocol or for tissue collection protocol, I think the patient should definitely enroll for that.   

Because that really helps our research strategy. But in terms of treatment, I think they should ask about available clinical trials that they qualify for.  

Katherine:

Let’s turn to clinical trials then. Patient participation, of course, is essential to finding new and better treatments. What would you say to someone who’s hesitant to participate in a clinical trial?  

Dr. Sen:

Yes. I mean, that’s often the thing. We hear about these novel drugs. They’re in trial. For a disease that’s that aggressive, I think once there is a relapse, I think clinical trials could be a very good option for patients. These are novel drugs that have come out of very robust research that we do in the lab. They can often work quite a bit. So, I think, of course, talk to your physicians. Talk to them at length about whether you do qualify for it. But if there is a trial at the center that you’re getting treated at and if the doctor advises that, I think enrolling in a clinical trial could be a very good option for patients, especially in the aggressive setting where there are not many options available for patients.  

As I mentioned here, research is my true north. I mean, all my lab does is understanding the biology of small cell. It’s extremely essential that we actually try to get the knowledge of the patient tumor. So, if you have availability of contributing either in terms of tissue or blood to research, I think I would advise and encourage patients to definitely contribute to that.